WO1996011013A1 - Cdr-grafted type iii anti-cea humanized mouse monoclonal antibodies - Google Patents

Cdr-grafted type iii anti-cea humanized mouse monoclonal antibodies Download PDF

Info

Publication number
WO1996011013A1
WO1996011013A1 PCT/US1995/011964 US9511964W WO9611013A1 WO 1996011013 A1 WO1996011013 A1 WO 1996011013A1 US 9511964 W US9511964 W US 9511964W WO 9611013 A1 WO9611013 A1 WO 9611013A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
cdr
antibody
region
monoclonal antibody
Prior art date
Application number
PCT/US1995/011964
Other languages
French (fr)
Inventor
Hans J. Hansen
Kathryn L. Armour
Original Assignee
Immunomedics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=23236916&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1996011013(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Immunomedics, Inc. filed Critical Immunomedics, Inc.
Priority to CA2200868A priority Critical patent/CA2200868C/en
Priority to EP95935020A priority patent/EP0783313B1/en
Priority to JP51258896A priority patent/JP3437580B2/en
Priority to DE69535755T priority patent/DE69535755D1/en
Priority to AU37196/95A priority patent/AU689331C/en
Publication of WO1996011013A1 publication Critical patent/WO1996011013A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3007Carcino-embryonic Antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6853Carcino-embryonic antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the invention relates to immunological reagents for diagnostic and therapeutic use in colon and other cancers.
  • the invention relates to humanized anti-carcinoembryonic antigen ("CEA”) monoclonal antibodies (“mAbs”) that have the binding affinity characteristics of corresponding mouse anti-CEA mAb (MN14) and the antigenic and effector properties of a human antibody.
  • CEA humanized anti-carcinoembryonic antigen
  • mAbs monoclonal antibodies
  • MN14 mouse anti-CEA mAb
  • the invention relates to humanized mAbs in which the complementarity determining regions ("CDRs") of an anti-CEA murine mAb is grafted into the framework regions of a human antibody, to DNAs that encode such CDR-grafted antibodies, to vectors and transformed hosts for propagating and expressing the DNAs, and to conjugates of the antibodies useful in diagnostic and therapeutic applications.
  • CDRs complementarity determining regions
  • circulating human anti- mouse antibodies HAMAs
  • HAMAs circulating human anti- mouse antibodies
  • circulating HAMAs decrease the effective concentration of the targeting agent in the patient and therefore limiting the diagnostic or therapeutic agent from reaching the target site.
  • One strategy has been to chemically modify the targeting antibody to suppress its antigenicity. For example, conjugation of polyethylene glycol to the targeting antibody (PEGylation) is reported to reduce antigenicity of antibodies.
  • Another approach has been to characterize the situs of antigenicity in an antibody and then remove it. In this vein, Fab', F(ab) 2 and other antibody fragments have been used in place of whole IgG.
  • attempts have been made to reduce the adverse effects of HAMA by plasmaphoretically removing HAMA from blood. Immunosuppressive techniques also have been used to ameliorate the adverse effect of the foreign antibody sufficiently to permit multiple treatments with the targeting agent.
  • Yet another object of the invention is to provide vectors for propagating the DNA and for expressing the antibody.
  • a related object of the invention is to provide cells containing a vector for the purposes of storage, propagation, antibody production and therapeutic applications.
  • Still another object of the invention is to provide compositions comprising the antibodies for use in diagnosis and therapy.
  • a humanized mouse mAb comprising the CDRs of a murine Class III, anti-CEA mAb (MN-14) engrafted to the framework regions of a heterologous (human) antibody, wherein the thus humanized mAb antibody retains the Class III, anti-CEA binding specificity of the murine mAb but in the patient is less immunogenic than is the parent MN-14 murine monoclonal antibody.
  • the light chain variable regions of the humanized antibody are characterized by the formula:
  • each FR is separately a framework region of a human antibody, and each CDR is separately in a complementarity-determining region of the light chains of MN-14, and the subscripts refer to light ("L") chain regions .
  • the heavy chain variable regions are characterized by the formula:
  • CDR L1 has the amino acid sequence KASQD VGTSVA (SEQ. ID NO. 20) ;
  • CDR ⁇ has the amino acid sequence WTSTR HT (SEQ. ID NO. 21) ;
  • CDR ⁇ has the amino acid sequence QQYSL YRS (SEQ. ID NO. 22) ;
  • CDR H1 has the amino acid sequence TY MS(SEQ. ID. NO.
  • CDR m has the amino acid sequence EIHP DSSTI NYAPS LKD (SEQ. ID NO. 24)
  • CDRr ⁇ has the amino acid sequence LYFGF PWFAY (SEQ. ID NO. 25) .
  • FR L1 has the amino acid sequence DIQLT QSPSS LSASV GDRVT ITC (SEQ. ID NO. 26) ; FR ⁇ has the amino acid sequence YQQK PGKAP KLLIY (SEQ. ID NO. 27) ; FR U has the amino acid sequence GVP(S or D) F SGS(G or V)S GTDFT FTISS QPED IATYY V (SEQ. ID NO. 28) ; FR has the amino acid sequence FGQGT KVIEK (SEQ. ID NO. 29) ; FR H1 has the amino acid sequence EVQLV ESGGG WQPG RS RL SCSSS GFDFT (SEQ. ID NO.
  • FR m has the amino acid sequence WVRQA PGKGL E VA (SEQ. ID NO. 33) , WVRQA PGKGL EWIA (SEQ. ID NO. 34) , or VRQP PGRGL EWIA (SEQ. ID NO. 35) ;
  • FRH 3 has the amino acid sequence RFTIS RDNSK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID NO.
  • FR H4 has the amino acid sequence WGQGT PVTVS S (SEQ. ID NO. 39), or WGQGT TVTVS S (SEQ. ID NO. 40); and wherein C may be in the sulfhydryl or disulfide form.
  • Another preferred embodiment comprises a diagnostic or therapeutic agent complexed to Class III, anti-CEA humanized mAb in which the CDRs of the antibody are derived from those of the MN-14 murine mAb and the FRs are derived from those of the heterologous (human) antibody, wherein the conjugate retains the Class III, anti-CEA binding specificity of MN-14, but is in humans less immunogenic than is murine MN-14.
  • the light chain and heavy chain variable regions are characterized as shown above and have amino acid sequences also as described above.
  • a method for diagnosing or treating a patient comprises the step of administering in an appropriate regimen the conjugate of the previous preferred embodiment.
  • Another preferred embodiment comprises an isolated, purified DNA that encodes the light chain, the heavy chain or both chains of the humanized antibody described above.
  • Another preferred embodiment comprises the DNA sequence of the CDRs and FRs described above.
  • CDR Complementarity Determining Region
  • FR is an abbreviation for Framework Region.
  • these are the portions of the variable regions of an antibody which lie adjacent to or flank the CDRs. In general, these regions have more of a structural function that affects the conformation of the variable region and are less directly responsible for the specific binding of antigen to antibody, although, nonetheless, the framework regions can affect the interaction.
  • Chimeric refers to an antibody in which the variable region is derived from a mouse antibody and the constant region is derived from an antibody from a heterologous (other) species.
  • Humanized refers to a chimeric antibody as defined above, but one in which the FR variable regions are derived from a human antibody.
  • HAMA refers to human antibodies directed to a mouse antibody, that are produced when a mouse antibody is administered to a human subject.
  • HAHA refers to human antibodies directed to a humanized mouse antibody.
  • CEA refers to carcinoembryonic antigen, a 180 kDa glycoprotein that is expressed in most adenocarcinomas of endodermally-derived digestive system epithelia and in some other cancers such as breast cancer and non-small cell lung cancer.
  • FIGURE 1 shows the consensus DNA sequence of murine NEWM MN-14 variable region heavy chain (“VH”) and its protein translation product. The CDRs are enclosed in boxes.
  • FIGURE 2 shows the consensus DNA sequence of murine MN-14 variable region light chain (“VK”) and its protein translation product. The CDRs are enclosed in boxes.
  • FIGURE 3 shows a vector for the expression of chimeric or humanized MN-14 heavy chain gene. The schematic diagram shows both the chimeric and a reshaped heavy chain immunoglobuiin (Ig) gene and the pSVgpt expression vector. The diagram at top, labeled "CHIMERIC, " is a map showing DNA encoding the MN-14 mouse VH region joined to DNA encoding a human IgGl constant region. In the MN-14 VH region the three CDRs are indicated by the three dark areas.
  • the FRs are indicated by the four stippled areas.
  • the middle diagram, labeled “RESHAPED” shows the humanization of the MN-14 VH region in which the mouse FRs have been replaced by human FRs, indicated by the four clear areas in the "Human” VH region.
  • the circular map of the expression vector pSVgpt at bottom shows the HindiII/BamHI insertion site for the reshaped MN-14 antibody gene just downstream from an Igh enhancer element.
  • the map also indicates some important functional domains in the vector, including the replication origins for propagation in E. coli (colEl ori) and in mammalian cells (SV40 promoter region) , and genes encoding selective markers for culturing bacterial
  • FIGURE 4 shows a vector for the expression of chimeric or humanized MN-14 kappa chain gene.
  • the diagram shows a chimeric and a reshaped MN-14 kappa light chain gene and the pSVhyg expression vector.
  • the diagram at top, labelled "CHIMERIC,” is a map showing DNA encoding the MN-14 mouse kappa light chain variable region ("VK") joined to DNA encoding a human kappa constant (“CK”) region.
  • VK mouse kappa light chain variable region
  • CK human kappa constant
  • the middle diagram shows the humanization of the MN-14 VK region in which the mouse FRs have been replaced by human FRs, which are indicated by the four clear areas in the "Human" VK region.
  • the circular map of the expression vector pSVhyg at bottom shows the HindiII/BamHI insertion site for the reshaped MN-14 antibody gene just downstream from an Igh enhancer element.
  • the map indicates some important functional domains in the vector, including the replication origins for E. coli ( col El ori ) and mammalian propagation (SV40 promoter region) , and genes encoding selective markers for culturing bacterial (Ap r ) and mammalian (grpt) cells transformed with the vector.
  • FIGURE 5 shows the alignments of the murine MN-14 variable regions (SEQ. ID. NOS. 2 and 4) with the human variable regions NEWM VH (SEQ. ID NO. 5) and REI VK (SEQ. ID NO. 6) ( Figure 5A) . and with the human KOL VH region (SEQ. ID NO. 7, ( Figure 5B) .
  • CDRs are boxed, and the murine VH FRs, which are incorporated into the humanized VH, are marked with their positions according to the numbering system of Kabat et al .
  • FIGURE 6 shows a comparison of the amino acid sequence between murine (SEQ. ID NO. 2) and humanized (SEQ. ID NOS. 5, 8-11, 7 and 12-15) .
  • MN-14 VH framework residues (FR) MN-14 VH framework residues (FR) . Only human FR residues different from the mouse are shown.
  • CDAs for NEWM and KOL are also not shown.
  • the areas of amino acid substitutions in the respective FRs are highlighted in bold, and the position of the substitution indicated according to the Kabat et al . numbering system.
  • the 3 CDRs are boxed.
  • FIGURE 7 shows the DNA sequence and corresponding amino acid sequence (SEQ. ID NOS. 16 and 17) of the MN- 14HuV ⁇ region. CDRs are boxed.
  • FIGURE 8 shows the DNA sequence and corresponding amino acid sequence (SEQ. ID NOS. 18 and 19) of the MN- 14HuVK region. CDRs are boxed.
  • FIGURE 9 is a graph of MN-14 blocking (i.e., competition) assays comparing relative binding affinities of KLHuVH variants, including: (-o- ) KLHuVH/HuVK; (- ⁇ -) KLHuVHAIG/HuVK; (-•-) KLHuVKAIGAY/HuVK; (- ⁇ -) KLHuVHAIGA/HuVK; (-A-) chimeric control; and, (- ⁇ -) murine control .
  • FIGURE 10 shows MN-14 blocking assays comparing hMN- 14 with HMN-14-NVT (glycosylated in FR 1 region) .
  • FIGURE 11 is a radioautogram of the abdomen of a colon cancer patient following administration of 131 I- labelled hMN14IgG (left panel) or mMN14IgG (right panel) in the same patient.
  • MN-14 The murine anti-CEA IgGl monoclonal antibody MN-14, and its production, have been described previously. Hansen et al . , Cancer, 71 : 3478 (1993) ; Primus et al . , U.S. Patent No. 4,818,709. MN-14 meets all of the criteria of a Class III, anti-CEA monoclonal antibody, being unreactive with meconium antigen by EIA and not reacting with normal tissues.
  • binding of humanized MN-14 may be assessed relative to a labeled MN- 14 probe.
  • a typical probe is MN-14 conjugated to horse radish peroxidase (HRP) .
  • HRP horse radish peroxidase
  • Both labeled and unlabeled MN- 14 are added to a CEA sample fixed to a solid support such as microtitre plate wells.
  • the degree of "blocking" of labeled MN-14 binding to CEA is a direct reflection of unlabeled MN-14 activity.
  • standard MN-14 the relative activity of an unknown sample of humanized MN-14 or derivatives thereof can be determined.
  • the reactions are performed in the wells of a microtitre plate where wells are charged directly with CEA at a level of, for example, 25 ⁇ g/well or indirectly where the wells are precharged with an antibody reactive with CEA but to an epitope different than that to which MN-14 interacts; such an antibody may be the MN-15 mAb.
  • CEA can thus be indirectly fixed to the well.
  • a competitive binding EIA assay can then be performed with such a charged plate.
  • antibodies can be radioiodinated conventionally with, for example, 131 I by the chloramine-T method to a specific activity of about 10 mCi/ ⁇ g, and free radioisotopes removed by chromatography on acrylamide gel columns (see Hansen et al . , 1993, above) .
  • Molecular biological techniques suitable to carrying out the invention as herein described also are known to those skilled in the art. Suitable teachings are described in numerous manuals and primary publications, /11013
  • MN-14 light and heavy chain CDRs disclosed herein, and modified MN-14 CDRs can be integrated into other antibodies using well-known recombinant techniques, such as those described in the above references.
  • oligonucleotides encoding MN-14 CDRs can be synthesized. Oligonucleotides that encode modified CDRs may be made, as well as those that encode exactly the amino acid sequences herein set forth. Also, the oligonucleotides may contain nucleotides in addition to those of an MN-14 CDRs, to facilitate cloning, for instance. Oligonucleotide synthesis techniques are well known, and can be carried out on automated equipment available from a number of manufacturers. Moreover, oligonucleotides of any specified sequence can be obtained commercially.
  • Oligonucleotides encoding the MN-14 CDRs and/or specific FR residues, or representing the complementary strand thereof may be used to introduce codons for these residues into VH or VK DNA by site-directed mutagenesis provided that the ends of the oligonucleotides, generally 12 nucleotides, are designed to anneal perfectly to the template DNA.
  • the template DNAs are typically single- stranded DNAs representing M13 vectors that carry a variable region DNA encoding the required FRs.
  • the mutagenic oligonucleotides are phosphorylated at their 5' ends and, together with an oligonucleotide priming 5' to variable region DNA, are annealedd to the ssDNA template.
  • the oligonucleotides are extended usinh T7 polymerase and the fragments linked together by T4 DNA ligase to give a complete mutant strand covering the whole variable region.
  • T4 DNA ligase to give a complete mutant strand covering the whole variable region.
  • multiple copies of its complementary strand can be synthesized from a suitable primer using Taq DNA polymerase in a thermal cycling reaction. Once the mutant strand has been preferentially amplified in this manner, the DNA can be amplified by conventional PCR for cloning, sequencing and expression.
  • Suitable antibody-encoding DNAs are illustrated by the disclosure herein, but include practically any such DNA.
  • a variety of human antibody genes are available in the form of publically accessible deposits. Many sequences of antibodies and antibody-encoding genes have been published and suitable antibody genes can be synthesized from these sequences much as described above.
  • the scope of this invention encompasses all alleles, variants and mutations of the DNA sequences described herein. CDR grafting in accordance with the present disclosure may be carried out using established
  • Antibody-producing cell lines may be selected and cultured using techniques well known to the skilled artisan. Such techniques are described in a variety of laboratory manuals and primary publications. For instance, techniques suitable for use in the invention as described below are described in CURRENT PROTOCOLS IN IMMUNOLOGY, Coligan et al . , Eds., Green Publishing Associates and Wiley-Interscience, John Wiley and Sons, New York (1991) which is herein incorporated by reference in its entirety, including supplements.
  • RNA may be isolated from the original hybridoma cells by standard techniques, such as guanidinium isothiocyanate extraction and precipitation followed by centrifugation or chromatography. Where desirable, mRNA may be isolated from total RNA by standard techniques such as chromatography on oligodT cellulose. Techniques suitable to these purposes are well known in the art as described in the foregoing references.
  • cDNAs that encode the light and the heavy chains of the antibody may be made, either simultaneously or separately, using reverse transcriptase and DNA polymerase in accordance with well known methods. It may be initiated by consensus constant region primers or by more specific primers based on the published heavy and light chain DNA and amino acid sequences.
  • PCR also may be used to isolate DNA clones encoding the antibody light and heavy chains.
  • the libraries may be screened by consensus primers or larger homologous probes, such as mouse constant region probes.
  • the necessary techniques are well known to those of skill in the art, are set forth in the foregoing Sambrook and Ausubel references and are illustrated by the examples set forth below.
  • cDNAs that encode the light and the heavy chain of an antibody can be propagated in any suitable vector in any suitable host prior to isolation of the CDR. Often the clones will most conveniently be propagated for this purpose in E. coli as illustrated in the examples below.
  • a variety of other vectors and host cells well knowN to those of skill profitably may be employed in this aspect of the invention. A variety of such vectors are described in the foregoing references.
  • DNA typically plasmid DNA
  • DNA may be isolated from the cells, restriction mapped and sequenced in accordance with standard, well known techniques set forth in detail in the foregoing references relating to recombinant DNA techniques.
  • DNAs encoding antibody heavy and light chains and fragments thereof in accordance with the vector are used to construct chimeric and CDR-grafted humanized MN-14 antibodies.
  • the CDRs of the MN-14 anti-CEA mAb are herein identified and described, and illustrated in Figures 1 and 2 (SEQ. ID NOS. 2 and 4, respectively) Using these sequences, CDRs of the MN-14 heavy and light chain can be synthesized for use in the present invention. It is not necessary to redone MN-14 CDRs from a natural source.
  • the DNA and amino acid sequences are set forth herein. Oligonucleotide synthesis techniques suitable to this aspect of the invention are well known to the skilled artisan and may be carried out using any of several commercially available automated synthesizers. In addition, DNAs encoding the CDRs set forth herein can be obtained through the services of commercial DNA synthesis vendors.
  • Polynucleotides synthesized in accordance with this aspect of the invention may include those not derived from an MN-14 CDR as well as those that make up the CDR.
  • the additional bases may be included to facilitate joining the CDR to the FRs from a heterologous source. They may comprise restriction sites or overlapping complementary regions for this purpose.
  • the synthesis of longer, double-stranded DNAs from shorter, overlapping, single-stranded DNAs is well known to those of skill in the art.
  • well known is the end-to-end joining of DNAs, including blunt-ended DNAs and those with at least partially overlapping complementary termini.
  • the CDRs of the MN-14 heavy and light chains may also be modified particularly after incorporation into a chimeric or humanized antibody using well-known recombinant DNA techniques for deleting, inserting and altering bases in a cloned or synthetic DNA or RNA.
  • Site-specific mutagenesis techniques suitable to this end are well known to those of skill in the art, and are illustrated in the foregoing references on recombinant DNA techniques.
  • deletional and insertional techniques are also illustrated. These methods can be used to introduce practically any desired alteration into polynucleotides that encode the MN-14 CDRs or into other regions of a closed heavy or light chain gene.
  • MN-14 CDRs and modified MN-14 CDRs can be introduced into practically any set of FRs in accordance with the present invention. It will be appreciated by those of skill in the art that a variety of well known techniques for cloning and manipulating polynucleotides may be effectively employed in this regard. Such techniques are illustrated by the methods set forth in the foregoing recombinant DNA-related references.
  • MN-14 CDRs are grafted into a human antibody.
  • human antibody in this context refers to any antibody that occurs in a human or an engineered antibody that has been designed, in some respect, to be compatible with the human immune system. Particularly preferred for this purpose are antibodies that, broadly, do not engender an adverse immune response in a patient. More particularly, the expression "human antibody” is intended to mean an antibody encoded by a gene actually occurring in a human, or an allele, variant or mutant thereof.
  • DNA encoding an MN-14-derived CDR-grafted antibody may be assembled from MN-14 VH and VK region DNAs and the variable regions thus formed combined with their respective light and heavy chains of human constant domains, it may be inserted into a vector for propagation and expression by conventional techniques. In this manner desired amounts of the antibody may be obtained.
  • the MN-14 CDR-grafted human antibody can be used in imaging applications by administrating to a subject the humanized antibody or Fab' thereof conjugated with an imaging comppund or isotope.
  • the antibody is conjugated to a label for imaging using conventional methods.
  • conventional methods include, but are not restricted to: 1) direct radioiodination of the antibody protein or fragments thereof or 2) direct attachment to the antibody or fragments thereof of metallic nuclides (see, e.g., Hansen et al . . Cancer, 73: 761 (1994)).
  • bifunctional chelates that can be used to bind various diagnostic or therepeutic metals to the antibody or fragment thereof is also within the scope of the present invention (see, aA ⁇ tiJbodies in Radiodiagnosis and Therapy, ed. M.R. Zalutsky, 1989, CRC Press, Boca Raton, FL, and Cancer Therapy wi th Radiolabeled Antibodies, ed.
  • the reaction of serum antibody with the MN-14 CDR- grafted antibody and imaging agent portions of the conjugate can be determined over the course of the diagnostic procedures, including the reaction of control sera obtained prior to administration of conjugate. Similar determinations are made in other patients treated with similar conjugates of MN-14 itself.
  • the sera antibody reactive with CDR-grafted MN-14 human antibodies detected by these tests is much less than the antibody reactive with antibody portion of the conjugate in patients treated with the murine MN-14-containing conjugates.
  • MN-14 and CDR-grafted MN-14 antibodies can be prepared as set forth above for conjugation to an aminodextran-boron adduct.
  • Amino-dextran-boron adducts can be prepared by reaction of a suitable boron cage compound (e.g., a 12- boron carborane suitably derivatized with an amino- dectran functional group) .
  • the amino-dextran is reacted with an excess of a haloacetyl acid ester or anhydride (such as iodoacetic anhydride) , thereby producing an amino-dextran with a desired number of haloacetyl groups, usually ranging from 10-1000 groups, depending on the reaction conditions and the size of the amino-dextran.
  • a suitable boron derivative such as mercaptocarborane-B12 is reacted, in a desired molar excess, with the haloacetyl-amino-dextran via an alkylation reaction.
  • a number of haloacetyl groups on the boronated haloacetyl amino-dextran remain unreacted, and can be used as a "handle" to attach the adduct to protein thiol groups.
  • MN-14 CDR-grafted humanized antibodies and their derivatives because of their reduced immunogenicity, are useful in therapy, for passive immunization without negative immune reactions such as serum sickness or anaphylactic shock, for localization and in vivo imaging of tumors as described above, for specific treatment of disease cells, e.g., site directed delivery of cytotoxins, immunomodulators or other pharmaceutically active molecules where local concentration of the active agent is an important factor, or the like, thereby establishing the practical utility of these humanized antibodies.
  • the humanized, CDR-grafted MN-14 monoclonal antibody is radiolabeled or conjugated with a metal chelator complexed with a radionuclide, e.g., iodine, ytrium, technetium, or the like, and radio-scanning techniques may be used to detect primary and metastatic CEA tumors.
  • a radionuclide e.g., iodine, ytrium, technetium, or the like
  • radio-scanning techniques may be used to detect primary and metastatic CEA tumors.
  • the radioactive antibody is injected, e.g., intravenously, and the patient scanned with a gamma imager at regular intervals. Tumors expressing CEA will take up more radioantibodies than other tissues and will be easily recognized by the imaging camera.
  • monoclonal antibodies labelled with 131 I are used in amounts of 3 to 10 ⁇ g representing 15 to 30 ⁇ Ci per kg body weight.
  • the antibodies are conjugated to any of a variety of known therapeutic agents such as doxorubicin, methotrexate, taxol, ricin A, radioactive atoms, cytoxic agents, and the like, formulating such conjugate in a pharmaceutically acceptable sterile vehicle, and administering the formulation by conventional means.
  • the therapeutic dosages can be readily determined conventionally by the user of average skills in these arts.
  • the therapeutic dose for mammals is between about 1 mg and 5 mg per kg body weight for the monoclonal antibodies themselves, and between 0.1 mg and 5 mg per kg body weight for conjugates with cytotoxic drugs, depending on the status of the patient and the mode of administration.
  • the humanized antibodies can be used in combination with components of the host's immune system, e.g., complement or cell mediated responses, in order to remove from the subject CEA- presenting cancer cells.
  • the immune responses of patients may be monitored in accordance with the foregoing procedures. For additional procedures for radioimaging and therapy, see EP 0 323,806, Hansen et al . , Cancer 71: 3478-85 (1993) , and U.S. Patent No. 4,818,709 and references contained therein, all of which are incorporated by reference.
  • compositions for parenteral administration such as are described in
  • a humanized antibody conjugate and a pharmaceutically acceptable carrier are administered to a patient in a therapeutically effective amount .
  • a combination of a conjugate and a pharmaceutically acceptable carrier is said to be administered in a "therapeutically effective amount” if the amount administered is physiologically significant.
  • An agent is “physiologically significant” if its presence results in a detectable change in the physiology of a recipient patient.
  • a targeted therapeutic agent is "therapeutically effective” if it delivers a higher proportion of the administered dose to the intended target than accretes at the target upon systemic administration of the equivalent untargeted agent.
  • the conjugate and carrier may need to be administered in combination with other therapeutic agents or as part of a broader treatment regimen.
  • Physicians are currently of the opinion that the effectiveness of targeted therapeutics can often be greatly increased when used in a combination therapy approach.
  • high-dose radioimmunotherapy for B-cell lymphomas which causes severe he atologic toxicity when used alone, has been shown to be highly effective when used in combination with autologous bone marrow reinfusion. Press et al . , "Treatment of Relapsed B Cell Lymphomas with High Dose Radioimmunotherapy and Bone Marrow Transplantation" in
  • the antibody was covalently complexed to aminodextran-conjugated methotrexate using the method described by Shih et al . , above using carbohydrate hydroxyl groups for derivatization purposes.
  • a mutation can be introduced at position 18-20 in the VK FRl region of hMN-14 (the prefix "h” is intended to mean “humanized”) so as to introduce a glycosylation site, NVT, prior to expression of the blocking gene in mammalian cells.
  • the Schiff bases are then reduced by treatment with excess NaBH 4
  • the amine-derivatized dextran then is purified by gel-exclusion chromatography.
  • the cytotoxic drug methotrexate (MTX) is activated by treatment with dicyclocarbodiamide, followed by reaction with N-hydroxysuccinimide, both in dimethylformamide. Activated MTX is mixed in a 50:1 ratio with the amino derivatized dextran in aqueous solution.
  • the product provides, after purification, MTX- derivatized dextran having about 35 MTX moles per mole.
  • the MTX adduct thus obtained is conjugated to a MN-14 CDR-grafted antibody using methods described in Shih, et al . , supra .
  • the antibody carbohydrates are oxidized and the resultant aldehydes are reacted with the remaining amines on the dextran in the adduct.
  • the Schiff-base product obtained thereby is reduced by treatment with sodium cyanoborohydride in 10-fold molar excess over antibody.
  • the reduced antibody-dextran-MTX product is thoroughly purified prior to assay, and formulated for administration to patients.
  • Parental MN-14 antibody is conjugated to dextran-MTX in the same way, as a control.
  • the purified CDR-grafted antibody conjugate can be administered to patients with a CEA-producing cancer (see above) .
  • the response to therapy is monitored, including adverse side effects, particularly those which are mediated by the patient's immune systems.
  • Patients treated with the CDR-grafted antibody conjugate show improved therapeutic results, decreased immune response to the agent and notably decreased immune-mediated adverse effects of therapy.
  • Therapy with the CDR-grafted antibody conjugate can be carried out at higher dosages and for longer periods of time then with the parental murine MN-14 antibody, allowing more aggressive therapies and improved responses.
  • Culturin ⁇ Antibody Producer Cells A mouse/mouse hybridoma cell line producing Class
  • anti-CEA monoclonal antibodies was established according to Hansen et al. (1993) above and Primus et al .
  • Cells were selected for secretion of kappa IgGl by testing conditioned medium using standard isotyping techniques. A variety of kits for this purpose are commercially available. Such cells were screened for production of antibody by testing conditioned medium using a standard blocking assay described above. Stocks of producer cells that proved out in the assay were expanded and frozen in liquid nitrogen.
  • MN-14-producing cells were expanded in culture, collected by centrifugation and washed. Total RNA was isolated from the cells in the pellet according to
  • EXAMPLE 3 cDNA Synthesis And Amplification Of The Heavy Chain Variable Region mRNA from MN-14 producing cells was used to synthesize cDNA using standard techniques of cDNA synthesis and DNA amplification by PCR, as described below.
  • the primers used for PCR included a restriction endonuclease cleavage site at their 5' ends to facilitate cloning of the amplification product.
  • the sequence of this primer, CG1FOR is shown in Table 1. Table 1 below provides other oligonucleotide sequences used herein.
  • VH1F0R 42 5' TGAGGAGACGGTGACCGTGGTCCCTTGGCCCCAG 3
  • SH1BACK 44 5' TGGAATTCATGGRATGGAGCTGGRTCWTBHTCTT 3'
  • SH2BACK 45 5' TGGAATTCATGRACTTCDGGYTCAACTKRRTTT 3 '
  • CK2FOR 46 5' GGAAGCTTGAAGATGGATACAGTTGGTGCAGC 3'
  • VK1F0R 47 55'' GTTAGATCTCCAGCTTGGTCCC 3'
  • VK3FOR 48 5' GTTAGATCTCCAGTTTGGTGCCT 3'
  • VK1BACK 49 5' GACATTCAGCTGACCCAGTCTCCA 3'
  • VK5BACK 53 5' TTGAATTCGGTGCCAGAKCWSAHATYGTKATG 3'
  • VK7BACK 55 5' TTGAATTCGGAGCTGATGGGAACATTGTAATG 3'
  • variable region of the heavy chain (“VH”) cDNA then was amplified by the PCR using the same primer,
  • VHIBACK a primer based on the consensus sequence of the 5' end of VH genes
  • the PCR product of this reaction was analyzed by agarose gel electrophoresis, which, upon ethidium bromide staining and fluorescence illumination, revealed one major band of about 400 bp, as expected.
  • signal sequence primers were used in the PCR to allow determination of the authentic amino acids of the N-terminus.
  • SH1BACK and SH2BACK degenerate oligonucleotides based on heavy chain signal sequence coding regions, were used in separate reactions in concert with CG1F0R.
  • a diffuse product band was obtained from CG1FOR, SH1BACK amplification.
  • VHIBACK primer pair was digested with HindiII and PstI separately. The cleavage sites of these enzymes are included in the PCR primers. It was preferable to determine whether there were also sites internal to the VH. Agarose gel analysis of the restriction fragments indicated the presence of an internal PstI site close to one end of the DNA.
  • the PCR product was digested with Hindlll and PstI, cloned into M13mpl8 and 19 and the DNA sequence of the inserts of representative clones determined. The majority of the clones contained inserts of the same VH DNA. The sequencing confirmed the presence of this additional, unexpected PstI site, which was close to the 3' end of the sequence of the CG1FOR primer partially encoding the final two amino acids of the VH.
  • VH clones were obtained by this method, further PCR product DNA was cloned as Pstl-PstI fragments. These clones thus contained full VH sequences but none of the constant region given by CG1FOR. A total of 16 full-length clones were obtained from the VK1BACK, CG1FOR product. In these experiments about 25% of the clones that were analyzed contained inserts unrelated to the VH region.
  • VH1FOR and SH1BACK primers In order to confirm the VH sequence from a second cDNA preparation and, at the same time, to obtain the authentic, rather than primer-dictated, DNA sequence corresponding to the N-terminus of the VH, the PCR product from VH1FOR and SH1BACK primers was cloned. These primers contain BstEII and EcoRI restriction sites, rather than the PstI and Hindlll sites of CG1FOR and VHIBACK described above. The PCR product of this reaction was cloned by digesting with BstEII, filling in the BstEII ends, digesting with EcoRI, and ligating the EcoRI and blunt ends to the vector, which had been digested with EcoRI and Hindll.
  • the sequences of the cloned fragments were determined. The yield of VH fragments was relatively low, perhaps reflecting a lack of specificity in the PCR caused by degeneracy of SH1BACK. However, four of the 18 clones that were sequenced contained DNA encoding the MN-14 VH region as previously sequenced. The other inserts did not derive from VH-encoding DNA.
  • the MN-14 CDR sequences are different from any of those reported by Kabat et al . (1987) , supra . Furthermore, the amino acids at four positions in the MN- 14 heavy chain VH framework regions are different from those in the framework regions of other subgroup IIIB VH sequences. These four substitutions (Ser 14, Thr 30, Ser 94 and Pro 108) have been observed in other murine VH regions outside the IIIB VH subgroup, however, with the proline in Kabat position 108 being the most unusual. Any unusual residues in the VH or VK may represent somatic mutations which proved adsvantageous to the binding of murine MN-14.
  • EXAMPLE 6 cDNA Synthesis And Amplification Of DNA Encoding
  • the MN-14 VK cDNA encoding the kappa light chain of MN-14 was cloned in much the same fashion as the cDNA encoding the variable region of the heavy chain, as described above.
  • CK2FOR The sequence of one primer, CK2FOR, was derived from the sequence of the 5' end of the constant region of kappa light chain genes ("CK") .
  • VKIFOR and VK3F0R were based on the sequence of the 3' end of the variable region of kappa light chain genes ("VK”) .
  • the first strand DNA product was amplified by PCR using a number of primer pairs. Synthesis in one direction was primed by the primers used to make the first strand. Polymerization in the other, "backward,” direction initiated from a series of kappa light chain- specific primers which had sequences based on either the sequence at the 5' end of the VK region, VK1BACK, VK2BACK, VK3BACK, VK4BACK and VK8BACK, or the sequence encoding the last four amino acids of the signal peptide and the first four amino acids of the variable region, VK5BACK, VK6BACK and VK7BACK.
  • cDNA primed by CK2FOR also was amplified using VK1F0R and VK8BACK.
  • the amplification products were analyzed by gel electrophoresis in the manner described in the Examples above.
  • the products from the reactions primed by VK1BACK, VK3BACK, VK5BACK, VK7BACK and VK8BACK gave rise to the expected 350 bp band.
  • EXAMPLE 7 Cloning and Sequencing the MN-14 Kavva Light Chain Variable Region Obtained by PCR Selected PCR products were cloned into M13mpl8 and 19 using the restriction sites included in the amplification primers in a manner similar to that described for VH in Example 4 above. Nucleotide sequencing revealed that most inserts were not VK- related. This is not uncommon when attempting to clone VK cDNAs and it appears to be more difficult to design VK-specific primers than VH specific primers.
  • VK1FOR/VK8BACK From the VK1FOR/VK8BACK combination, a VK cDNA insert was obtained, but this did not yield a functional VK due to a frameshift within the cDNA encoding CDR L3 and absence of the invariant Cys at position 23.
  • This VK cDNA has been isolated from other hybridoma cells and it is derived from the Sp2/0 fusion partner.
  • CK2FOR/VK1BACK product yielded a further four different aberrant VK cDNA inserts, in this case lacking the conserved residues of framework 4.
  • VK8BACK which does not extend as far into the VK gene.
  • analysis of further clones from VK1FOR/VK8BACK product did not yield the desired insert.
  • VK3FOR was designed from its genuine fourth framework sequence, and synthesized as an alternative to VK1FOR.
  • VK framework regions (Met 10, Val 66 and Thr 76) do not appear in other members of this subgroup. Met 10 and Val
  • variable region cDNAs In the production of a chimeric antibody consisting of murine variable regions and human constant regions, testing alongside the parent mouse antibody served to check that the correct variable region cDNAs have been isolated. A successful chimeric antibody also acts as a useful control when assessing the binding of humanized versions.
  • the scheme used in cloning the variable regions for expression is described by Orlandi et al .
  • VH DNA was amplified from the M13 clone MNVH41 using the PCR with oligonucleotides VHIBACK and VH1FOR.
  • the PstI and BstEII restriction sites in the primers allowed the VH to be inserted into M13VHPCR1 in the correct context for expression.
  • the internal BatriHI restriction site of the VH was removed by site directed mutagenesis.
  • the reaction product, which encompassed the entire Hindlll-BamHI fragment of M13VHPCR1 was cloned into pSVgpt and the VH sequence confirmed.
  • the human IgGl constant region gene published by Takahashi et al . , Cell , 29: 671-679 (1962) above then was added to the construct as a B-at ⁇ HI fragment, which yielded the vector referred to as pSVgptMN14MuVHHuIgGl.
  • VK DNA was similarly obtained from the M13 clone MNVK154 by PCR amplification with the primers VK8BACK and VK3FOR and the PvuII, Bglll-digested product cloned into M13VKPCR1, whereupon the sequence of the variable region was checked.
  • the HindiII-BamHI fragment was excised from RF DNA and transferred to the plasmid pSVhyg.
  • the construct already contains a human kappa constant region gene as described in Hieter et al . , Cell , 22: 197-207 (1980) .
  • the final vector thus obtained was designated pSVhygMN14MuVKHuCK.
  • the purified antibody was characterized further with reference to native MN-14 antibody, human antibodies and other controls.
  • the antibody was also characterized by its reaction profile in a MN-14 blocking assay, which provided an informative comparison of CEA binding affinities of the hybrid antibodies with CEA binding by the MN-14 murine antibody which served as a positive control. 8.3 Humanization of the MN-14 antibody
  • the human NEWM VH, KOL VH and REI VK frameworks were chosen as the basis for reshaping the antibody, as they are likely to be tolerated in humans. Alignments of the MN-14 VH (SEQ. ID NO. 2) and VK (SEQ. ID NO. 4) with these human variable regions are shown in Figure 5 (SEQ. ID. NOS. 5-7) .
  • the starting points for the introduction of MN-14 CDRs are DNAs encoding the required FRs and irrelevant CDRs. These template variable regions are in a form compatable with the expression vectors used, that is, within HindiII-BamHI fragments, that also include promotor regions, signal peptide and intron DNA ( Figures 3 and 4) .
  • the template is M13VHPCR1 (Orlandi et al , above, and section 8.3 below) .
  • a derivative of this template, containing KOL FRs and irrelevant CDRs, was used to generate the KOL coding region.
  • a derivative of M13VKPCR1 (Orlandi et ai. above) was used in the creation of the HuVK vector.
  • the resulting vectors were termed M13NMHuVH, M13KLHuVH and M13HuVK.
  • the HindiII-BamHI fragments containing the humanized MN-14 variable region DNAs were transferred from these M13 vectors to the expression vectors essentially as described for the construction of the chimeric MN-14 expression vectors in Example 8.1.
  • the NEWM FR is described in Poljak et al . Biochemistry 16: 3412-20 (1977) . Construction of a hMN- 14 with an affinity for CEA comparable to that of its murine counterpart was achieved in a stepwise approach. Production of the chimeric antibody provided a useful control when assessing the binding of the humanized versions.
  • the human NEWM VH and REI VK frameworks were initially chosen as the basis for reshaping the antibody as they are known to be tolerated in man. MN-14 residues Phe27, Asp28 and Thr30 were retained because, although not part of the Rabat's hypervariable region, CDR1 residues 31-35, those amino acids are part of the CDR1 structural loop (Chothia et al .
  • KOL VH is described in Schmidt et al . , Z. physiol . Chem, 364: 713-747 (1987) .
  • the FR used for humanizing the antibody is as given in Kabat.
  • the original version of the KOL-based VH contained the
  • the humanized KLHuVH varients such as antibody KLHuVHAIGA/HuVK, were purified and tested in a blocking assay carried out as follows. Antibodies were added at the indicated concentrations together with HRP-labeled MN-14 to a final volume of 0.1 ml. Following 30 mins. of incubation at 37 C C, and washing to remove unbound antibodies, the relative affinities of the antibodies were determined from the remaining bound peroxidase activity. A shown by the assay data of Figure 9, the activity of the "reshaped" (i.e., CDR-engrafted on FR) humanized antibody was similar to that of chimeric and murine positive controls.
  • REI VL is described in Epp et al . , Eur. J. Biochem. , 45: 513-24 (1974). No change4s were made in REI framework to improve binding.
  • the changes from the sequences given in Kabat et al. (1987) are: M4 to L; T39 to K: Y71 to F; L104 to V; Q105 to E: and T107 to K. These changes preexisted in the template used when grafting the MN-14 CDRs and were not made specifically to improve binding.
  • the M4 to L change incorporates a restriction site but the remaining differences eliminate unusual residues in the REI framework.
  • a similar framework has been referred to as a consensus of huma kappa subgroup I by Foote et al. , J. Mol . Biol . 224: 487- 9 (1992) EXAMPLE 9
  • MN-14 CDR-grafted Humanized Antibodies Cells that were stably transformed for expression of the MN-14 CDR-grafted human antibodies were selected in the manner described above and cloned out to establish individual producer lines. Each of the lines was assayed to determine production of the correct antibody and to assess the efficiency of production. The antibody class was determined and the anti-CEA binding affinity assessed. The best producers were further characterized for the overall amount of the antibody produced and, for the best of these, sequences were obtained from the mRNA to insure that the mutation has not occurred in the antibody genes during transfection, integration, propagation or selection.
  • the best producer lines of the MN-14 CDR-grafted human antibody were cultured, the growth medium collected and filtered through a 0.2 micron membrane.
  • the antibody was then purified by protein A chromatography followed by other conventional purification steps such as ion exchange and size exclusion chromotography.
  • the cells were pelleted and from the supernatant by conventional centrifugation.
  • the antibody was purified from the supernatant fluid as described above.

Abstract

A humanized monoclonal antibody, comprising the complementarity-determining regions of a parental murine Class III, anti-CEA monoclonal antibody engrafted to the framework regions of a heterologous antibody, wherein the humanized monoclonal antibody retains the binding specificity of, but is less immunogenic in a heterologous host than, the parental murine monoclonal antibody. A preferred murine Class III, anti-CEA monoclonal antibody is the MN-14 antibody and the preferred heterologous antibody is from a human. Also provided are DNA constructs and vectors for producing the humanized monoclonal antibodies, and diagnostic and therapeutic conjugates using same.

Description

CDR-GRAFTED TYPE III ANTI-CEA HUMANIZED MOUSE MONOCLONAL ANTIBODIES
BACKGROUND OF THE INVENTION
The invention relates to immunological reagents for diagnostic and therapeutic use in colon and other cancers. In particular, the invention relates to humanized anti-carcinoembryonic antigen ("CEA") monoclonal antibodies ("mAbs") that have the binding affinity characteristics of corresponding mouse anti-CEA mAb (MN14) and the antigenic and effector properties of a human antibody. Further, the invention relates to humanized mAbs in which the complementarity determining regions ("CDRs") of an anti-CEA murine mAb is grafted into the framework regions of a human antibody, to DNAs that encode such CDR-grafted antibodies, to vectors and transformed hosts for propagating and expressing the DNAs, and to conjugates of the antibodies useful in diagnostic and therapeutic applications.
A promising approach to cancer diagnosis and therapy involves the use of targeting antibodies to deliver diagnostic and therapeutic agents directly to the site of a malignancy. Over the past decade, a wide variety of tumor-specific antibodies and antibody fragments have been developed, as have methods to conjugate the antibodies to drugs, toxins, radionuclides or other agents, and to administer the conjugates to patients. These efforts have produced great progress, but a variety of largely unanticipated problems have limited the diagnostic and therapeutic utility of some of the reagents thus far developed.
Among the most intractable problems is that which is caused by the human immune system itself, which may respond to the targeting conjugate as a foreign antigen. For instance, patients treated with drugs or 1
radionuclides complexed with murine monoclonal antibodies (which have been the most commonly used targeting antibodies for human) develop circulating human anti- mouse antibodies (HAMAs) and a generalized immediate type-III hypersensitivity reaction to the antibody moiety of the conjugate. Furthermore, even when adverse side effects are minimal (for example, as in a single administration) , circulating HAMAs decrease the effective concentration of the targeting agent in the patient and therefore limiting the diagnostic or therapeutic agent from reaching the target site.
Several approaches have been developed to overcome or avoid this problem, with only limited success. One strategy has been to chemically modify the targeting antibody to suppress its antigenicity. For example, conjugation of polyethylene glycol to the targeting antibody (PEGylation) is reported to reduce antigenicity of antibodies. Another approach has been to characterize the situs of antigenicity in an antibody and then remove it. In this vein, Fab', F(ab)2 and other antibody fragments have been used in place of whole IgG. In addition, attempts have been made to reduce the adverse effects of HAMA by plasmaphoretically removing HAMA from blood. Immunosuppressive techniques also have been used to ameliorate the adverse effect of the foreign antibody sufficiently to permit multiple treatments with the targeting agent.
None of these approaches has proven altogether satisfactory. An important need persists for a means to reduce or eliminate the adverse immune response to targeting antibody and antibody conjugates in order to gain the full benefit of these diagnostic and therapeutic agents.
This goal has been achieved with the CDR-grafted humanized murine anti-human CEA mAbs that are described below. SUMMARY OF THE INVENTION It is an object of the present invention to provide a humanized Class III anti-CEA mAb in which the CDRs of a murine Class III anti-CEA mAb (MN14) are functionally engrafted to the amino acid sequence of a human antibody or antibody fragment to provide an immunological reagent with the anti-CEA binding properties of the murine Class III, anti-CEA mAb and the immunogenic properties of a human mAb in a human patient. It is another object of the present invention to provide DNA constructs encoding such antibodies. Particular objects in this regard are substrate DNAs that facilitate genetic manipulation to produce improved antibodies and DNAs encoding the antibodies with advantageous properties in cell culture and antibody production.
Yet another object of the invention is to provide vectors for propagating the DNA and for expressing the antibody. A related object of the invention is to provide cells containing a vector for the purposes of storage, propagation, antibody production and therapeutic applications.
Still another object of the invention is to provide compositions comprising the antibodies for use in diagnosis and therapy. In this regard it is an object of the invention to provide conjugates comprising the antibodies complexed with imaging agents and therapeutic agents for ex vivo and in vivo imaging, diagnosis, prognosis and therapy, among others. In accomplishing the foregoing objects, there has been provided, in accordance with one aspect of the present invention, a humanized mouse mAb, comprising the CDRs of a murine Class III, anti-CEA mAb (MN-14) engrafted to the framework regions of a heterologous (human) antibody, wherein the thus humanized mAb antibody retains the Class III, anti-CEA binding specificity of the murine mAb but in the patient is less immunogenic than is the parent MN-14 murine monoclonal antibody. In a highly preferred embodiment, the light chain variable regions of the humanized antibody are characterized by the formula:
FRL1-CDRL1-FR^-CDRLJ-FRU-CDRL3-FRU wherein each FR is separately a framework region of a human antibody, and each CDR is separately in a complementarity-determining region of the light chains of MN-14, and the subscripts refer to light ("L") chain regions . The heavy chain variable regions are characterized by the formula:
FRH1-CDRH]-FRjc-CDRrø-FRrø-CDRH3-FRH4 wherein FR and CDR have the same meanings as above, and wherein the subscripts "H" refer to heavy chain regions. In one embodiment, CDRL1 has the amino acid sequence KASQD VGTSVA (SEQ. ID NO. 20) ; CDR^ has the amino acid sequence WTSTR HT (SEQ. ID NO. 21) ; CDR^ has the amino acid sequence QQYSL YRS (SEQ. ID NO. 22) ; CDRH1 has the amino acid sequence TY MS(SEQ. ID. NO. 23) ; CDRm has the amino acid sequence EIHP DSSTI NYAPS LKD (SEQ. ID NO. 24) ; and, CDRrø has the amino acid sequence LYFGF PWFAY (SEQ. ID NO. 25) .
In another embodiment, FRL1 has the amino acid sequence DIQLT QSPSS LSASV GDRVT ITC (SEQ. ID NO. 26) ; FR^ has the amino acid sequence YQQK PGKAP KLLIY (SEQ. ID NO. 27) ; FRU has the amino acid sequence GVP(S or D) F SGS(G or V)S GTDFT FTISS QPED IATYY V (SEQ. ID NO. 28) ; FR has the amino acid sequence FGQGT KVIEK (SEQ. ID NO. 29) ; FRH1 has the amino acid sequence EVQLV ESGGG WQPG RS RL SCSSS GFDFT (SEQ. ID NO. 30) , EVQLV ESGGG WQPG RSLRL SCSAS GFDFT (SEQ. ID NO. 31) , or QVQLQ ESGPG LVRPS QTLSL TCTSS GFDFT (SEQ. ID NO. 32) ; FRm has the amino acid sequence WVRQA PGKGL E VA (SEQ. ID NO. 33) , WVRQA PGKGL EWIA (SEQ. ID NO. 34) , or VRQP PGRGL EWIA (SEQ. ID NO. 35) ; FRH3 has the amino acid sequence RFTIS RDNSK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID NO. 36) , RFTIS RDNAK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID NO. 37) , or RVTML RDTSK NGSFL RLSSV TAADT AVYYC AS (SEQ. ID NO. 38) ; and 6/11013 PC17US95/11964
- 5 -
FRH4 has the amino acid sequence WGQGT PVTVS S (SEQ. ID NO. 39), or WGQGT TVTVS S (SEQ. ID NO. 40); and wherein C may be in the sulfhydryl or disulfide form.
Another preferred embodiment comprises a diagnostic or therapeutic agent complexed to Class III, anti-CEA humanized mAb in which the CDRs of the antibody are derived from those of the MN-14 murine mAb and the FRs are derived from those of the heterologous (human) antibody, wherein the conjugate retains the Class III, anti-CEA binding specificity of MN-14, but is in humans less immunogenic than is murine MN-14. In one such embodiment the light chain and heavy chain variable regions are characterized as shown above and have amino acid sequences also as described above. In yet another preferred embodiment, a method for diagnosing or treating a patient comprises the step of administering in an appropriate regimen the conjugate of the previous preferred embodiment.
Another preferred embodiment comprises an isolated, purified DNA that encodes the light chain, the heavy chain or both chains of the humanized antibody described above.
Another preferred embodiment comprises the DNA sequence of the CDRs and FRs described above. Other objects, features and advantages of the present invention will become apparent from the following detailed description and appended claims.
ILLUSTRATIVE GLOSSARY
The following terms or abbreviations are used in the present application. The meanings set out in this glossary are for illustrative purposes only. The full meaning of the terms will be apparent to those of skill in the art.
"CDR" is used as an abbreviation for Complementarity Determining Region. These are the regions within the variable regions of an antibody that are primarily, but not exclusively, responsible for antigen-antibody binding.
"FR" is an abbreviation for Framework Region. Broadly speaking, these are the portions of the variable regions of an antibody which lie adjacent to or flank the CDRs. In general, these regions have more of a structural function that affects the conformation of the variable region and are less directly responsible for the specific binding of antigen to antibody, although, nonetheless, the framework regions can affect the interaction.
"Chimeric" refers to an antibody in which the variable region is derived from a mouse antibody and the constant region is derived from an antibody from a heterologous (other) species.
"Humanized" refers to a chimeric antibody as defined above, but one in which the FR variable regions are derived from a human antibody.
"HAMA" refers to human antibodies directed to a mouse antibody, that are produced when a mouse antibody is administered to a human subject.
"HAHA" refers to human antibodies directed to a humanized mouse antibody.
"CEA" refers to carcinoembryonic antigen, a 180 kDa glycoprotein that is expressed in most adenocarcinomas of endodermally-derived digestive system epithelia and in some other cancers such as breast cancer and non-small cell lung cancer.
The letter "h" as a prefix means "humanized" . Other abbreviations are used in accordance with Roitt et al . , IMMUNOLOGY, 3rd ed. Mosby Year Book Europe Ltd. (1993) , the entirety of which is herein incorporated by reference.
These and other terms used in the present disclosure are used in the same sense as ordinarily they are employed in the arts to which this invention pertains. BRIEF DESCRIPTION OF THE FIGURES
FIGURE 1 (SEQ. ID. NOS. 1 and 2) shows the consensus DNA sequence of murine NEWM MN-14 variable region heavy chain ("VH") and its protein translation product. The CDRs are enclosed in boxes.
FIGURE 2 (SEQ. ID. NOS. 3 and 4) shows the consensus DNA sequence of murine MN-14 variable region light chain ("VK") and its protein translation product. The CDRs are enclosed in boxes. FIGURE 3 shows a vector for the expression of chimeric or humanized MN-14 heavy chain gene. The schematic diagram shows both the chimeric and a reshaped heavy chain immunoglobuiin (Ig) gene and the pSVgpt expression vector. The diagram at top, labeled "CHIMERIC, " is a map showing DNA encoding the MN-14 mouse VH region joined to DNA encoding a human IgGl constant region. In the MN-14 VH region the three CDRs are indicated by the three dark areas. The FRs are indicated by the four stippled areas. The middle diagram, labeled "RESHAPED" shows the humanization of the MN-14 VH region in which the mouse FRs have been replaced by human FRs, indicated by the four clear areas in the "Human" VH region. The circular map of the expression vector pSVgpt at bottom shows the HindiII/BamHI insertion site for the reshaped MN-14 antibody gene just downstream from an Igh enhancer element. The map also indicates some important functional domains in the vector, including the replication origins for propagation in E. coli (colEl ori) and in mammalian cells (SV40 promoter region) , and genes encoding selective markers for culturing bacterial
(Apr) and mammalian (gpt) cells transformed with the vector. Expression of the antibody gene in this case is mediated by the Ig promoter indicated by the solid circles near the Hindi11 site in the maps of the antibody genes.
FIGURE 4 shows a vector for the expression of chimeric or humanized MN-14 kappa chain gene. The diagram shows a chimeric and a reshaped MN-14 kappa light chain gene and the pSVhyg expression vector. The diagram at top, labelled "CHIMERIC," is a map showing DNA encoding the MN-14 mouse kappa light chain variable region ("VK") joined to DNA encoding a human kappa constant ("CK") region. In the mouse VK region the three CDRs are indicated by the three dark areas and the FRs are indicated by the four stippled areas. The middle diagram, labelled "RESHAPED" shows the humanization of the MN-14 VK region in which the mouse FRs have been replaced by human FRs, which are indicated by the four clear areas in the "Human" VK region. The circular map of the expression vector pSVhyg at bottom shows the HindiII/BamHI insertion site for the reshaped MN-14 antibody gene just downstream from an Igh enhancer element. The map indicates some important functional domains in the vector, including the replication origins for E. coli ( col El ori ) and mammalian propagation (SV40 promoter region) , and genes encoding selective markers for culturing bacterial (Apr) and mammalian (grpt) cells transformed with the vector. Expression of the antibody gene in this case is mediated by the Ig promoter schematized by the solid circles near the Hindi11 site in the maps of the antibody genes. FIGURE 5 shows the alignments of the murine MN-14 variable regions (SEQ. ID. NOS. 2 and 4) with the human variable regions NEWM VH (SEQ. ID NO. 5) and REI VK (SEQ. ID NO. 6) (Figure 5A) . and with the human KOL VH region (SEQ. ID NO. 7, (Figure 5B) . CDRs are boxed, and the murine VH FRs, which are incorporated into the humanized VH, are marked with their positions according to the numbering system of Kabat et al . SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, U.S. Government Printing Office, Washington, D.C., 1987. Murine residues outside the CDRs that were included in the KLHuVH are indicated by a filled circle.
FIGURE 6 shows a comparison of the amino acid sequence between murine (SEQ. ID NO. 2) and humanized (SEQ. ID NOS. 5, 8-11, 7 and 12-15) . MN-14 VH framework residues (FR) . Only human FR residues different from the mouse are shown. CDAs for NEWM and KOL are also not shown. The areas of amino acid substitutions in the respective FRs are highlighted in bold, and the position of the substitution indicated according to the Kabat et al . numbering system. The 3 CDRs are boxed.
FIGURE 7 shows the DNA sequence and corresponding amino acid sequence (SEQ. ID NOS. 16 and 17) of the MN- 14HuVΗ region. CDRs are boxed.
FIGURE 8 shows the DNA sequence and corresponding amino acid sequence (SEQ. ID NOS. 18 and 19) of the MN- 14HuVK region. CDRs are boxed.
FIGURE 9 is a graph of MN-14 blocking (i.e., competition) assays comparing relative binding affinities of KLHuVH variants, including: (-o- ) KLHuVH/HuVK; (-■-) KLHuVHAIG/HuVK; (-•-) KLHuVKAIGAY/HuVK; (-□-) KLHuVHAIGA/HuVK; (-A-) chimeric control; and, (-Δ-) murine control . FIGURE 10 shows MN-14 blocking assays comparing hMN- 14 with HMN-14-NVT (glycosylated in FR 1 region) .
FIGURE 11 is a radioautogram of the abdomen of a colon cancer patient following administration of 131I- labelled hMN14IgG (left panel) or mMN14IgG (right panel) in the same patient.
DETAILED DESCRIPTION OF THE INVENTION
Notwithstanding past failures to develop an effective non-HAMA-inducing anti-CEA antibody having the CEA- binding characteristics of MN-14, it has been discovered that the CDRs of the MN-14 mAb can be grafted onto the
FRs of a human antibody to provide antibodies and antibody-derived reagents that have the antigen binding properties of the MN-14 anti-CEA mAb, while also exhibiting reduced induction of HAMA and augmented effector activities.
The murine anti-CEA IgGl monoclonal antibody MN-14, and its production, have been described previously. Hansen et al . , Cancer, 71 : 3478 (1993) ; Primus et al . , U.S. Patent No. 4,818,709. MN-14 meets all of the criteria of a Class III, anti-CEA monoclonal antibody, being unreactive with meconium antigen by EIA and not reacting with normal tissues.
Blocking studies are carried out according to Hansen et al . 1993, above, Losman et al . , Int. Cancer, 56 : 580
(1994) ; Hansen et al . , Clin . Chem. , 35: 146 (1989) .
Using the same conditions as described in those references for quantification of CEA, binding of humanized MN-14 may be assessed relative to a labeled MN- 14 probe. A typical probe is MN-14 conjugated to horse radish peroxidase (HRP) . Both labeled and unlabeled MN- 14 are added to a CEA sample fixed to a solid support such as microtitre plate wells. The degree of "blocking" of labeled MN-14 binding to CEA is a direct reflection of unlabeled MN-14 activity. Using standard MN-14, the relative activity of an unknown sample of humanized MN-14 or derivatives thereof can be determined. Typically, the reactions are performed in the wells of a microtitre plate where wells are charged directly with CEA at a level of, for example, 25 μg/well or indirectly where the wells are precharged with an antibody reactive with CEA but to an epitope different than that to which MN-14 interacts; such an antibody may be the MN-15 mAb. CEA can thus be indirectly fixed to the well. A competitive binding EIA assay can then be performed with such a charged plate.
Alternate to the aforementioned HRP-labeled mAbs, antibodies can be radioiodinated conventionally with, for example, 131I by the chloramine-T method to a specific activity of about 10 mCi/μg, and free radioisotopes removed by chromatography on acrylamide gel columns (see Hansen et al . , 1993, above) . Molecular biological techniques suitable to carrying out the invention as herein described also are known to those skilled in the art. Suitable teachings are described in numerous manuals and primary publications, /11013
- 11 -
including inter alia, Sambrook et al . , MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989) , PROTOCOLS IN MOLECULAR BIOLOGY, Ausubel et al . , Eds., Green Publishing Associates and Wiley-Interscience, John Wiley and Sons, New York (1987, 1988, 1989), which are herein incorporated by reference in their entirety including supplements.
MN-14 light and heavy chain CDRs disclosed herein, and modified MN-14 CDRs can be integrated into other antibodies using well-known recombinant techniques, such as those described in the above references.
Specific methods suitable to this end are shown below in the examples. Based on the amino acid sequences set forth herein, oligonucleotides encoding MN-14 CDRs can be synthesized. Oligonucleotides that encode modified CDRs may be made, as well as those that encode exactly the amino acid sequences herein set forth. Also, the oligonucleotides may contain nucleotides in addition to those of an MN-14 CDRs, to facilitate cloning, for instance. Oligonucleotide synthesis techniques are well known, and can be carried out on automated equipment available from a number of manufacturers. Moreover, oligonucleotides of any specified sequence can be obtained commercially.
Oligonucleotides encoding the MN-14 CDRs and/or specific FR residues, or representing the complementary strand thereof, may be used to introduce codons for these residues into VH or VK DNA by site-directed mutagenesis provided that the ends of the oligonucleotides, generally 12 nucleotides, are designed to anneal perfectly to the template DNA. The template DNAs are typically single- stranded DNAs representing M13 vectors that carry a variable region DNA encoding the required FRs. In one method, the mutagenic oligonucleotides are phosphorylated at their 5' ends and, together with an oligonucleotide priming 5' to variable region DNA, are annealedd to the ssDNA template. The oligonucleotides are extended usinh T7 polymerase and the fragments linked together by T4 DNA ligase to give a complete mutant strand covering the whole variable region. Using the mutant strand as a template, multiple copies of its complementary strand can be synthesized from a suitable primer using Taq DNA polymerase in a thermal cycling reaction. Once the mutant strand has been preferentially amplified in this manner, the DNA can be amplified by conventional PCR for cloning, sequencing and expression. Suitable antibody-encoding DNAs are illustrated by the disclosure herein, but include practically any such DNA. A variety of human antibody genes are available in the form of publically accessible deposits. Many sequences of antibodies and antibody-encoding genes have been published and suitable antibody genes can be synthesized from these sequences much as described above. The scope of this invention encompasses all alleles, variants and mutations of the DNA sequences described herein. CDR grafting in accordance with the present disclosure may be carried out using established
-techniques. Antibody-producing cell lines may be selected and cultured using techniques well known to the skilled artisan. Such techniques are described in a variety of laboratory manuals and primary publications. For instance, techniques suitable for use in the invention as described below are described in CURRENT PROTOCOLS IN IMMUNOLOGY, Coligan et al . , Eds., Green Publishing Associates and Wiley-Interscience, John Wiley and Sons, New York (1991) which is herein incorporated by reference in its entirety, including supplements.
RNA may be isolated from the original hybridoma cells by standard techniques, such as guanidinium isothiocyanate extraction and precipitation followed by centrifugation or chromatography. Where desirable, mRNA may be isolated from total RNA by standard techniques such as chromatography on oligodT cellulose. Techniques suitable to these purposes are well known in the art as described in the foregoing references. cDNAs that encode the light and the heavy chains of the antibody may be made, either simultaneously or separately, using reverse transcriptase and DNA polymerase in accordance with well known methods. It may be initiated by consensus constant region primers or by more specific primers based on the published heavy and light chain DNA and amino acid sequences. PCR also may be used to isolate DNA clones encoding the antibody light and heavy chains. In this case the libraries may be screened by consensus primers or larger homologous probes, such as mouse constant region probes. The necessary techniques are well known to those of skill in the art, are set forth in the foregoing Sambrook and Ausubel references and are illustrated by the examples set forth below. cDNAs that encode the light and the heavy chain of an antibody can be propagated in any suitable vector in any suitable host prior to isolation of the CDR. Often the clones will most conveniently be propagated for this purpose in E. coli as illustrated in the examples below. However, a variety of other vectors and host cells well knowN to those of skill profitably may be employed in this aspect of the invention. A variety of such vectors are described in the foregoing references.
DNA, typically plasmid DNA, may be isolated from the cells, restriction mapped and sequenced in accordance with standard, well known techniques set forth in detail in the foregoing references relating to recombinant DNA techniques.
DNAs encoding antibody heavy and light chains and fragments thereof in accordance with the vector are used to construct chimeric and CDR-grafted humanized MN-14 antibodies.
The CDRs of the MN-14 anti-CEA mAb are herein identified and described, and illustrated in Figures 1 and 2 (SEQ. ID NOS. 2 and 4, respectively) Using these sequences, CDRs of the MN-14 heavy and light chain can be synthesized for use in the present invention. It is not necessary to redone MN-14 CDRs from a natural source. The DNA and amino acid sequences are set forth herein. Oligonucleotide synthesis techniques suitable to this aspect of the invention are well known to the skilled artisan and may be carried out using any of several commercially available automated synthesizers. In addition, DNAs encoding the CDRs set forth herein can be obtained through the services of commercial DNA synthesis vendors.
Polynucleotides synthesized in accordance with this aspect of the invention may include those not derived from an MN-14 CDR as well as those that make up the CDR. The additional bases may be included to facilitate joining the CDR to the FRs from a heterologous source. They may comprise restriction sites or overlapping complementary regions for this purpose. The synthesis of longer, double-stranded DNAs from shorter, overlapping, single-stranded DNAs is well known to those of skill in the art. Likewise, well known is the end-to-end joining of DNAs, including blunt-ended DNAs and those with at least partially overlapping complementary termini. These techniques are illustrated in the foregoing references on recombinant DNA techniques, for instance.
The CDRs of the MN-14 heavy and light chains may also be modified particularly after incorporation into a chimeric or humanized antibody using well-known recombinant DNA techniques for deleting, inserting and altering bases in a cloned or synthetic DNA or RNA. Site-specific mutagenesis techniques suitable to this end are well known to those of skill in the art, and are illustrated in the foregoing references on recombinant DNA techniques. Also illustrated are deletional and insertional techniques. These methods can be used to introduce practically any desired alteration into polynucleotides that encode the MN-14 CDRs or into other regions of a closed heavy or light chain gene. MN-14 CDRs and modified MN-14 CDRs can be introduced into practically any set of FRs in accordance with the present invention. It will be appreciated by those of skill in the art that a variety of well known techniques for cloning and manipulating polynucleotides may be effectively employed in this regard. Such techniques are illustrated by the methods set forth in the foregoing recombinant DNA-related references.
In a particularly preferred embodiment of the present invention, MN-14 CDRs are grafted into a human antibody. It will be understood that human antibody in this context refers to any antibody that occurs in a human or an engineered antibody that has been designed, in some respect, to be compatible with the human immune system. Particularly preferred for this purpose are antibodies that, broadly, do not engender an adverse immune response in a patient. More particularly, the expression "human antibody" is intended to mean an antibody encoded by a gene actually occurring in a human, or an allele, variant or mutant thereof.
Once DNA encoding an MN-14-derived CDR-grafted antibody has been assembled from MN-14 VH and VK region DNAs and the variable regions thus formed combined with their respective light and heavy chains of human constant domains, it may be inserted into a vector for propagation and expression by conventional techniques. In this manner desired amounts of the antibody may be obtained.
The MN-14 CDR-grafted human antibody can be used in imaging applications by administrating to a subject the humanized antibody or Fab' thereof conjugated with an imaging comppund or isotope.
The antibody is conjugated to a label for imaging using conventional methods. Such conventional methods include, but are not restricted to: 1) direct radioiodination of the antibody protein or fragments thereof or 2) direct attachment to the antibody or fragments thereof of metallic nuclides (see, e.g., Hansen et al . . Cancer, 73: 761 (1994)). The use of bifunctional chelates that can be used to bind various diagnostic or therepeutic metals to the antibody or fragment thereof is also within the scope of the present invention (see, aAπtiJbodies in Radiodiagnosis and Therapy, ed. M.R. Zalutsky, 1989, CRC Press, Boca Raton, FL, and Cancer Therapy wi th Radiolabeled Antibodies, ed. D.M. Goldenberg, 1994, CRC Press, Boca Raton, FL) . Following the conjugation procedure and characterization of the product, satisfactorily labelled conjugates are purified to homogeneity under conditions that conform to Good Manufacturing Procedures ("GMP") appropriate to the production of diagnostic compositions for use in human patients.
The reaction of serum antibody with the MN-14 CDR- grafted antibody and imaging agent portions of the conjugate can be determined over the course of the diagnostic procedures, including the reaction of control sera obtained prior to administration of conjugate. Similar determinations are made in other patients treated with similar conjugates of MN-14 itself. The sera antibody reactive with CDR-grafted MN-14 human antibodies detected by these tests is much less than the antibody reactive with antibody portion of the conjugate in patients treated with the murine MN-14-containing conjugates.
Humanized MN-14 antibodies conjugated to aminodextran and to boron may be used for diagnostic purposes. MN-14 and CDR-grafted MN-14 antibodies can be prepared as set forth above for conjugation to an aminodextran-boron adduct. Amino-dextran-boron adducts can be prepared by reaction of a suitable boron cage compound (e.g., a 12- boron carborane suitably derivatized with an amino- dectran functional group) . In a preferred embodiment, the amino-dextran is reacted with an excess of a haloacetyl acid ester or anhydride (such as iodoacetic anhydride) , thereby producing an amino-dextran with a desired number of haloacetyl groups, usually ranging from 10-1000 groups, depending on the reaction conditions and the size of the amino-dextran. A suitable boron derivative such as mercaptocarborane-B12 is reacted, in a desired molar excess, with the haloacetyl-amino-dextran via an alkylation reaction. In a preferred embodiment, a number of haloacetyl groups on the boronated haloacetyl amino-dextran remain unreacted, and can be used as a "handle" to attach the adduct to protein thiol groups.
MN-14 CDR-grafted humanized antibodies and their derivatives, because of their reduced immunogenicity, are useful in therapy, for passive immunization without negative immune reactions such as serum sickness or anaphylactic shock, for localization and in vivo imaging of tumors as described above, for specific treatment of disease cells, e.g., site directed delivery of cytotoxins, immunomodulators or other pharmaceutically active molecules where local concentration of the active agent is an important factor, or the like, thereby establishing the practical utility of these humanized antibodies. As described above, for in vivo imaging, the humanized, CDR-grafted MN-14 monoclonal antibody is radiolabeled or conjugated with a metal chelator complexed with a radionuclide, e.g., iodine, ytrium, technetium, or the like, and radio-scanning techniques may be used to detect primary and metastatic CEA tumors. To that end, the radioactive antibody is injected, e.g., intravenously, and the patient scanned with a gamma imager at regular intervals. Tumors expressing CEA will take up more radioantibodies than other tissues and will be easily recognized by the imaging camera. Preferentially, monoclonal antibodies labelled with 131I are used in amounts of 3 to 10 μg representing 15 to 30 μCi per kg body weight. For therapy with cytotoxic agents, the antibodies are conjugated to any of a variety of known therapeutic agents such as doxorubicin, methotrexate, taxol, ricin A, radioactive atoms, cytoxic agents, and the like, formulating such conjugate in a pharmaceutically acceptable sterile vehicle, and administering the formulation by conventional means. The therapeutic dosages can be readily determined conventionally by the user of average skills in these arts. The therapeutic dose for mammals is between about 1 mg and 5 mg per kg body weight for the monoclonal antibodies themselves, and between 0.1 mg and 5 mg per kg body weight for conjugates with cytotoxic drugs, depending on the status of the patient and the mode of administration. Alternately, the humanized antibodies can be used in combination with components of the host's immune system, e.g., complement or cell mediated responses, in order to remove from the subject CEA- presenting cancer cells. The immune responses of patients may be monitored in accordance with the foregoing procedures. For additional procedures for radioimaging and therapy, see EP 0 323,806, Hansen et al . , Cancer 71: 3478-85 (1993) , and U.S. Patent No. 4,818,709 and references contained therein, all of which are incorporated by reference.
Preferred are pharmaceutical preparations for parenteral administration, such as are described in
Remington's Pharmaceutical Sciences, Mack Publishing Co. , Easton, PA, 1989. The final preparations contain from 0.01% to 50% of active ingredients. Methods for the production of such conjugates and their use in diagnostics and therapeutics are provided in, for example, Shih et al. , U.S. Patent No. 5,057,313; Shih et al . , Int . J. Cancer 41:832 (1988) ; copending, commonly owned USSN 08/162,912; and, McKearn et al. , U.S. Patent No. 5,156,840, the contents of which are incorporated by reference.
As noted above, for purposes of therapy, a humanized antibody conjugate and a pharmaceutically acceptable carrier are administered to a patient in a therapeutically effective amount . A combination of a conjugate and a pharmaceutically acceptable carrier is said to be administered in a "therapeutically effective amount" if the amount administered is physiologically significant. An agent is "physiologically significant" if its presence results in a detectable change in the physiology of a recipient patient. A targeted therapeutic agent is "therapeutically effective" if it delivers a higher proportion of the administered dose to the intended target than accretes at the target upon systemic administration of the equivalent untargeted agent.
To be therapeutically effective the conjugate and carrier may need to be administered in combination with other therapeutic agents or as part of a broader treatment regimen. Physicians are currently of the opinion that the effectiveness of targeted therapeutics can often be greatly increased when used in a combination therapy approach. For example, high-dose radioimmunotherapy for B-cell lymphomas, which causes severe he atologic toxicity when used alone, has been shown to be highly effective when used in combination with autologous bone marrow reinfusion. Press et al . , "Treatment of Relapsed B Cell Lymphomas with High Dose Radioimmunotherapy and Bone Marrow Transplantation" in
CANCER THERAPY WITH RADIOLABELED ANTIBODIES, Goldenberg, ed. (CRC Press, Boca Raton, 1994) ch. 17. In another example a five-fold enhancement of tumor uptake of a radiolabeled antibody is observed when the tumor is preirradiated. Leichner et al . , Int . J. Radiat . Oncol . Biol . Phys . 14:1033 (1987). Mechanisms which have been shown to have the potential for improving the clinical efficacy of radioimmunotherapy are also discussed in DeNardo et al . , "Overview of Obstacles and Opportunities for Radioimmunotherapy of Cancer" in CANCER THERAPY WITH
RADIOLABELED ANTIBODIES, Goldenberg, Ed. (CRC Press, Boca Raton, 1994) ch. 11. Methods of developing such combination protocols, as well as to investigate dose- limiting side effects and to potentiate and amplify targeting, uptake, and beneficial side effects, are well known to skilled clinical artisans in this field and would not require undue experimentation to develop. In vivo experiments using conjugates of the humanized MN-14 with diagnostic and therapeutic agents have been carried out with animal models and with human patients (see Example 11 below) . The CDR-grafted humanized antibody conjugate exhibited a better therapeutic profile and could be used in longer treatment regimens than the parental MN-14 antibody conjugate. The CDR-grafted antibody conjugate provided a better therapeutic effect and fewer deleterious side effects than the control murine antibody conjugates.
For example, the antibody was covalently complexed to aminodextran-conjugated methotrexate using the method described by Shih et al . , above using carbohydrate hydroxyl groups for derivatization purposes. In order to determine the contribution of antibody carbohydrate groups on immunoactivity, a mutation can be introduced at position 18-20 in the VK FRl region of hMN-14 (the prefix "h" is intended to mean "humanized") so as to introduce a glycosylation site, NVT, prior to expression of the blocking gene in mammalian cells. Comparison of hMN-14 antibody with mutated hMN-14-NVT in a blocking cell binding assay (Figure 8) has demonstrated that the carbohydrate moiety at position 18 is without influence on immunoreactivity of this humanized antibody. Aminodextran, average molecular mass 40 kDa, is oxidized by NaI04 to form aldehydes (by the oxidation of hydroxyl groups) . About 50 to 150 moles of aldehydic groups are introduced per mole of aminodextran by careful control of the reaction conditions and timing. The aldehydes then are reacted with an excess of 1,3-diamino- 2-hydroxypropane to form Schiff bases with virtually all of the aldehydes. The Schiff bases are then reduced by treatment with excess NaBH4 The amine-derivatized dextran then is purified by gel-exclusion chromatography. The cytotoxic drug methotrexate (MTX) is activated by treatment with dicyclocarbodiamide, followed by reaction with N-hydroxysuccinimide, both in dimethylformamide. Activated MTX is mixed in a 50:1 ratio with the amino derivatized dextran in aqueous solution. The product provides, after purification, MTX- derivatized dextran having about 35 MTX moles per mole. The MTX adduct thus obtained is conjugated to a MN-14 CDR-grafted antibody using methods described in Shih, et al . , supra . For example, the antibody carbohydrates are oxidized and the resultant aldehydes are reacted with the remaining amines on the dextran in the adduct. The Schiff-base product obtained thereby is reduced by treatment with sodium cyanoborohydride in 10-fold molar excess over antibody. The reduced antibody-dextran-MTX product is thoroughly purified prior to assay, and formulated for administration to patients.
Parental MN-14 antibody is conjugated to dextran-MTX in the same way, as a control.
The purified CDR-grafted antibody conjugate can be administered to patients with a CEA-producing cancer (see above) . The response to therapy is monitored, including adverse side effects, particularly those which are mediated by the patient's immune systems. Patients treated with the CDR-grafted antibody conjugate show improved therapeutic results, decreased immune response to the agent and notably decreased immune-mediated adverse effects of therapy. Therapy with the CDR-grafted antibody conjugate can be carried out at higher dosages and for longer periods of time then with the parental murine MN-14 antibody, allowing more aggressive therapies and improved responses.
The present invention is further described by reference to the following, illustrative examples. It will be appreciated that the techniques related to isolating DNA clones encoding MN-14 light and heavy chain genes are illustrated by cloning techniques useful to isolate light and heavy chain genes of any antibody from producing cells. There is no necessity, given the disclosed sequence, to reisolate MN-14 heavy and light chain genes to carry out the invention. It should be understood that the detailed description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from the following illustrative description.
EXAMPLE 1
Culturinσ Antibody Producer Cells A mouse/mouse hybridoma cell line producing Class
III, anti-CEA monoclonal antibodies was established according to Hansen et al. (1993) above and Primus et al .
(1983) above.
Cells were selected for secretion of kappa IgGl by testing conditioned medium using standard isotyping techniques. A variety of kits for this purpose are commercially available. Such cells were screened for production of antibody by testing conditioned medium using a standard blocking assay described above. Stocks of producer cells that proved out in the assay were expanded and frozen in liquid nitrogen.
EXAMPLE 2
Isolating RNA From Producing Cell Lines
MN-14-producing cells were expanded in culture, collected by centrifugation and washed. Total RNA was isolated from the cells in the pellet according to
Favaloro et al . , Methods in Enzγmolocrγ 65 : 718 (1980) and
Orlandi et al . , Proc . Na t 'l Acad . Sci . , USA £36.: 3833
(1989) , which are incorporated by reference. EXAMPLE 3 cDNA Synthesis And Amplification Of The Heavy Chain Variable Region mRNA from MN-14 producing cells was used to synthesize cDNA using standard techniques of cDNA synthesis and DNA amplification by PCR, as described below. In general, the primers used for PCR included a restriction endonuclease cleavage site at their 5' ends to facilitate cloning of the amplification product. An oligonucleotide complementary to the end of the sense strand of the DNA encoding the first constant region domain of the murine IgGj heavy chain ("CHI") was used to prime first strand cDNA synthesis by reverse transcriptase. The sequence of this primer, CG1FOR, is shown in Table 1. Table 1 below provides other oligonucleotide sequences used herein.
TABLE 1
OLIGONUCLEOTIDE SEQUENCES
SEQ.
ID
NO,
CGIFOR 41 5' GGAAGCTTAGACAGATGGGGGTGTCGTTTTG 3'
VH1F0R 42 5' TGAGGAGACGGTGACCGTGGTCCCTTGGCCCCAG 3
VH1BACK 43 5' AGGTSMARCTGCAGSAGTCWGG 3'
SH1BACK 44 5' TGGAATTCATGGRATGGAGCTGGRTCWTBHTCTT 3'
SH2BACK 45 5' TGGAATTCATGRACTTCDGGYTCAACTKRRTTT 3 '
CK2FOR 46 5' GGAAGCTTGAAGATGGATACAGTTGGTGCAGC 3'
VK1F0R 47 55'' GTTAGATCTCCAGCTTGGTCCC 3'
VK3FOR 48 5' GTTAGATCTCCAGTTTGGTGCCT 3'
VK1BACK 49 5' GACATTCAGCTGACCCAGTCTCCA 3'
VK2BACK 50 5' GACRTTCAGCTGACCCAGGMTGMA 3'
VK3BACK 51 5' GACATTCAGCTGACCCA 3'
VK4BACK 52 5' GACATTGAGCTCACCCAGTCTCCA 3'
VK5BACK 53 5' TTGAATTCGGTGCCAGAKCWSAHATYGTKATG 3'
VK6BACK 54 5' TTGAATTCGGTGCCAGAKCWSAHATYGTKCTC 3'
VK7BACK 55 5' TTGAATTCGGAGCTGATGGGAACATTGTAATG 3'
VK8BACK 56 5' CWGAGAAATTCAGCTGACCCAGTCTC 3'
Restriction sites incorporated in primers to facilitate cloning are underlined.
The variable region of the heavy chain ("VH") cDNA then was amplified by the PCR using the same primer,
CG1FOR, and a primer based on the consensus sequence of the 5' end of VH genes (VHIBACK) , as described in Orlandi et al . (1989) cited above. The PCR product of this reaction was analyzed by agarose gel electrophoresis, which, upon ethidium bromide staining and fluorescence illumination, revealed one major band of about 400 bp, as expected. For confirmatory sequences from a second cDNA preparation, signal sequence primers were used in the PCR to allow determination of the authentic amino acids of the N-terminus. SH1BACK and SH2BACK, degenerate oligonucleotides based on heavy chain signal sequence coding regions, were used in separate reactions in concert with CG1F0R. A diffuse product band was obtained from CG1FOR, SH1BACK amplification.
In order to increase the VH content of the product it was excised from low melting point agarose and amplified using SH1BACK and an oligonucleotide complementary to a fourth framework region consensus sequence, VH1F0R. This product of this reaction was a discrete band when analyzed by agarose gel electrophoresis. EXAMPLE 4
Cloning And Sequencing DNA Encoding The MN-14 Heavy Chain Variable Region Obtained By PCR
The amplification product obtained using the CG1FOR,
VHIBACK primer pair was digested with HindiII and PstI separately. The cleavage sites of these enzymes are included in the PCR primers. It was preferable to determine whether there were also sites internal to the VH. Agarose gel analysis of the restriction fragments indicated the presence of an internal PstI site close to one end of the DNA. The PCR product was digested with Hindlll and PstI, cloned into M13mpl8 and 19 and the DNA sequence of the inserts of representative clones determined. The majority of the clones contained inserts of the same VH DNA. The sequencing confirmed the presence of this additional, unexpected PstI site, which was close to the 3' end of the sequence of the CG1FOR primer partially encoding the final two amino acids of the VH. Although 6/11013
- 25 -
several full length VH clones were obtained by this method, further PCR product DNA was cloned as Pstl-PstI fragments. These clones thus contained full VH sequences but none of the constant region given by CG1FOR. A total of 16 full-length clones were obtained from the VK1BACK, CG1FOR product. In these experiments about 25% of the clones that were analyzed contained inserts unrelated to the VH region.
In order to confirm the VH sequence from a second cDNA preparation and, at the same time, to obtain the authentic, rather than primer-dictated, DNA sequence corresponding to the N-terminus of the VH, the PCR product from VH1FOR and SH1BACK primers was cloned. These primers contain BstEII and EcoRI restriction sites, rather than the PstI and Hindlll sites of CG1FOR and VHIBACK described above. The PCR product of this reaction was cloned by digesting with BstEII, filling in the BstEII ends, digesting with EcoRI, and ligating the EcoRI and blunt ends to the vector, which had been digested with EcoRI and Hindll. The sequences of the cloned fragments were determined. The yield of VH fragments was relatively low, perhaps reflecting a lack of specificity in the PCR caused by degeneracy of SH1BACK. However, four of the 18 clones that were sequenced contained DNA encoding the MN-14 VH region as previously sequenced. The other inserts did not derive from VH-encoding DNA.
In all, 20 full-length MN-14 VH clones were obtained. Five transition mutations were observed amongst the sequences in the MN-14 VH region clones. These mutations are likely to have been introduced during amplification as a result of misincorporation by Tag polymerase.
EXAMPLE 5
Analysis Of The Amino Acid Sequence Of The Heavy Chain Variable Region Of MN-14
The amino acid sequence of murine MN-14 heavy chain variable region, translated from the VH DNA sequence, is shown in Figure 1 (SEQ. ID. NOS. 1 and 2) . Comparison of this sequence with sequences representing the murine VH subgroups indicated that the variable heavy region of MN- 14 belongs to subgroup IIIB (see, Kabat et al . SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST, U.S. Government Printing Office, 1987) .
The MN-14 CDR sequences are different from any of those reported by Kabat et al . (1987) , supra . Furthermore, the amino acids at four positions in the MN- 14 heavy chain VH framework regions are different from those in the framework regions of other subgroup IIIB VH sequences. These four substitutions (Ser 14, Thr 30, Ser 94 and Pro 108) have been observed in other murine VH regions outside the IIIB VH subgroup, however, with the proline in Kabat position 108 being the most unusual. Any unusual residues in the VH or VK may represent somatic mutations which proved adsvantageous to the binding of murine MN-14.
EXAMPLE 6 cDNA Synthesis And Amplification Of DNA Encoding The MN-14 VK cDNA encoding the kappa light chain of MN-14 was cloned in much the same fashion as the cDNA encoding the variable region of the heavy chain, as described above.
Several primers were used to prime reverse transcriptase for synthesis of the first strand of the kappa chain cDNA. The sequence of one primer, CK2FOR, was derived from the sequence of the 5' end of the constant region of kappa light chain genes ("CK") . The sequences of two other primers, VKIFOR and VK3F0R, were based on the sequence of the 3' end of the variable region of kappa light chain genes ("VK") .
The first strand DNA product was amplified by PCR using a number of primer pairs. Synthesis in one direction was primed by the primers used to make the first strand. Polymerization in the other, "backward," direction initiated from a series of kappa light chain- specific primers which had sequences based on either the sequence at the 5' end of the VK region, VK1BACK, VK2BACK, VK3BACK, VK4BACK and VK8BACK, or the sequence encoding the last four amino acids of the signal peptide and the first four amino acids of the variable region, VK5BACK, VK6BACK and VK7BACK. In addition, cDNA primed by CK2FOR also was amplified using VK1F0R and VK8BACK. The amplification products were analyzed by gel electrophoresis in the manner described in the Examples above. The products from the reactions primed by VK1BACK, VK3BACK, VK5BACK, VK7BACK and VK8BACK gave rise to the expected 350 bp band.
EXAMPLE 7 Cloning and Sequencing the MN-14 Kavva Light Chain Variable Region Obtained by PCR Selected PCR products were cloned into M13mpl8 and 19 using the restriction sites included in the amplification primers in a manner similar to that described for VH in Example 4 above. Nucleotide sequencing revealed that most inserts were not VK- related. This is not uncommon when attempting to clone VK cDNAs and it appears to be more difficult to design VK-specific primers than VH specific primers.
From the VK1FOR/VK8BACK combination, a VK cDNA insert was obtained, but this did not yield a functional VK due to a frameshift within the cDNA encoding CDRL3 and absence of the invariant Cys at position 23. This VK cDNA has been isolated from other hybridoma cells and it is derived from the Sp2/0 fusion partner. CK2FOR/VK1BACK product yielded a further four different aberrant VK cDNA inserts, in this case lacking the conserved residues of framework 4. A fifth VK insert obtained using this primer pair was that of a functional VK with the exception of a frameshift at the 3'end of VK1BACK, a phenomenon apparently due to mismatch-induced slippage of the primer. This problem may be avoided by the use of VK8BACK which does not extend as far into the VK gene. However, analysis of further clones from VK1FOR/VK8BACK product did not yield the desired insert. In order to amplify preferentially the putative VK,
VK3FOR was designed from its genuine fourth framework sequence, and synthesized as an alternative to VK1FOR.
This strategy proved successful when amplification of VK3FOR-primed cDNA with VK3FOR and VK8BACK yielded 4 clones containing the desired VK.
The DNA and amino acid sequuence of the murine MN-14 kappa light chain variable region is set out in Figure 2
(SEQUENCE ID. NOS. 3 and 4) . This MN-14 VK can be placed in Rabat's VK subgroup V. Only 3 residues in the MN-14
VK framework regions (Met 10, Val 66 and Thr 76) do not appear in other members of this subgroup. Met 10 and Val
66 are the most unusual of the three. They are not found in any murine VK listed in Kabat. None of the MN-14 VK CDRs are previosuly-reported sequences in Kabat.
EXAMPLE 8
Grafting of the MN-14 VH and VK CDRs into Variable Regions of Human Antibody
8.1. Construction of chomeric anitbodv expression vectors
In the production of a chimeric antibody consisting of murine variable regions and human constant regions, testing alongside the parent mouse antibody served to check that the correct variable region cDNAs have been isolated. A successful chimeric antibody also acts as a useful control when assessing the binding of humanized versions. The scheme used in cloning the variable regions for expression is described by Orlandi et al .
(1989) supra , and is illustrated in Figures 3 and 4. VH DNA was amplified from the M13 clone MNVH41 using the PCR with oligonucleotides VHIBACK and VH1FOR. The PstI and BstEII restriction sites in the primers allowed the VH to be inserted into M13VHPCR1 in the correct context for expression. At this point, the internal BatriHI restriction site of the VH was removed by site directed mutagenesis. The reaction product, which encompassed the entire Hindlll-BamHI fragment of M13VHPCR1 was cloned into pSVgpt and the VH sequence confirmed.
The human IgGl constant region gene, published by Takahashi et al . , Cell , 29: 671-679 (1962) above then was added to the construct as a B-atπHI fragment, which yielded the vector referred to as pSVgptMN14MuVHHuIgGl.
VK DNA was similarly obtained from the M13 clone MNVK154 by PCR amplification with the primers VK8BACK and VK3FOR and the PvuII, Bglll-digested product cloned into M13VKPCR1, whereupon the sequence of the variable region was checked. The HindiII-BamHI fragment was excised from RF DNA and transferred to the plasmid pSVhyg. The construct already contains a human kappa constant region gene as described in Hieter et al . , Cell , 22: 197-207 (1980) . The final vector thus obtained was designated pSVhygMN14MuVKHuCK.
8.2 Expression and testing of the hybrid antibody The HindiII-BamHI fragment of M13KLHuVHAIGA was inserted into a plasmid pSVgpt to yield the expression vector pSVgptKLHuAIGAHuIgGl. Similarly, the Hindlll - BamHI fragment of M13HuVK was inserted into the plasmid pSVhyg to yield the expression vector pSVhygMN14REIHuVKHuCK. About 5μg pSVgptMN14MuVHHuIgGl and lOμg pSVhygMN14MuVKHuCK DNAs were linearized with Pvul and transferred into about 107 subconfluent SP2/0 myeloma cells by electroporation conventionally using a BioRad Model 165BR1160 Gene Pulser Electroporator with a single pulse of 170 V, 960 μF. Cells were selected for the expression of the gpt gene in 24-well plates by addition of mycophenolic acid and xanthine to the DMEM + 10% FCS growth medium.
Wells which contained colonies of surviving cells were identified. The supernatant medium was removed from these wells and assayed for human antibody. C lonies that secreted antibodies were expanded to give 0.5 L of conditioned medium for isolation of larger amounts of antibody. Antibody was purified conventionally from the medium by protein-A agarose affinity chromatography, initially.
The purified antibody was characterized further with reference to native MN-14 antibody, human antibodies and other controls.
The antibody was also characterized by its reaction profile in a MN-14 blocking assay, which provided an informative comparison of CEA binding affinities of the hybrid antibodies with CEA binding by the MN-14 murine antibody which served as a positive control. 8.3 Humanization of the MN-14 antibody
The human NEWM VH, KOL VH and REI VK frameworks were chosen as the basis for reshaping the antibody, as they are likely to be tolerated in humans. Alignments of the MN-14 VH (SEQ. ID NO. 2) and VK (SEQ. ID NO. 4) with these human variable regions are shown in Figure 5 (SEQ. ID. NOS. 5-7) . A. NEWM based humanization.
The starting points for the introduction of MN-14 CDRs are DNAs encoding the required FRs and irrelevant CDRs. These template variable regions are in a form compatable with the expression vectors used, that is, within HindiII-BamHI fragments, that also include promotor regions, signal peptide and intron DNA (Figures 3 and 4) . For the NEWM VH version, the template is M13VHPCR1 (Orlandi et al , above, and section 8.3 below) . A derivative of this template, containing KOL FRs and irrelevant CDRs, was used to generate the KOL coding region. A derivative of M13VKPCR1 (Orlandi et ai. above) was used in the creation of the HuVK vector. The resulting vectors were termed M13NMHuVH, M13KLHuVH and M13HuVK. The HindiII-BamHI fragments containing the humanized MN-14 variable region DNAs were transferred from these M13 vectors to the expression vectors essentially as described for the construction of the chimeric MN-14 expression vectors in Example 8.1.
The NEWM FR is described in Poljak et al . Biochemistry 16: 3412-20 (1977) . Construction of a hMN- 14 with an affinity for CEA comparable to that of its murine counterpart was achieved in a stepwise approach. Production of the chimeric antibody provided a useful control when assessing the binding of the humanized versions. The human NEWM VH and REI VK frameworks were initially chosen as the basis for reshaping the antibody as they are known to be tolerated in man. MN-14 residues Phe27, Asp28 and Thr30 were retained because, although not part of the Rabat's hypervariable region, CDR1 residues 31-35, those amino acids are part of the CDR1 structural loop (Chothia et al . , J. Mol . Biol . 176: 901- 917 (1987) ) . In addition, the following residues were also selected for incorporation into the humanized VH for the following reasons: Ala24, this residue contacts CDR1; Arg7l, the side chain of this residue pokes through the center of the domain to interact with CDRs 1 and 2; substitution of the smaller Val may alter the conformation of these CDR loops; and, Ser94, the majority of antibodies have Arg in this position where it is thought to interact with an Asp residue on CDR3, and the inclusion of the Arg of NEWM could create an unwanted interaction with the murine CDR. Other changes were made to this version (NMHuVH) in three areas corresponding to regions which had been proved important in other reshaped molecules. These changes were:
(i) Gln77Phe78Ser79 to ThrLeuTyr (NMHuVhHTLY, SEQ.
ID NO. 9) (ii) Ser82Thr83Ala84Ala85 to LysArgSerGlu (NMHuVhHKRSE, SEQ. ID NO. 10) (iii) Arg66Val67Thr68Met69Leu70 to LysPhelleValSer
(NMHuVhKFIVS, SEQ. ID NO. 11) The alignment of the different versions of the NEWM VH frameworks (SEQ. ID NOS. 5 and 8-11) with the MN-14 VH (SEQ. ID NO. 2) is shown in Figure 6. Each of these versions has been paired with the same HuVK. The inclusion of either the TLY or KFIVS motifs gave about a two-fold improvement. There are 2 differences from the NEWN framework sequences given in Kabat et. al (1987) above: S107 to T and L108 to T. Kabat lists residue 1 as PCA and residues 5 and 6 as E or Q. B. KOL based humanization.
In parallel to the use of NEWM VH, we have also reshaped the human KOL VH. KOL VH is described in Schmidt et al . , Z. physiol . Chem, 364: 713-747 (1987) . The FR used for humanizing the antibody is as given in Kabat. The original version of the KOL-based VH contained the
MN-14 CDRs, as defined in terms of residue variability
(Kabat et al . , above) and three additional murine residues. As with the NEWM VH, two of these substitutions were made because the actual peptide CDR1 structural loop, which extends from the /3-sheet framework, consists of residues 25 to 32 (Chothia et al . 1987, above) . Changes at positions 28 and 30 allowed this loop to be transplanted as a whole from the murine antibody. MN-14 residue 94 was included because the Arg residue of the KOL VH is involved in a salt bridge with AspllO and, like with the NEWM VH, it was felt that retention of Arg 94 might perturb the MN-14 CDR3 structure. The side-chain of residue 94 may also interact with residues of CDR1. Other changes made to this basis MN-14 KOLHuVH (SEQ. ID NO. 12) were as follows: (i) Ser24 to Ala24 and Val48Ala49 to IleGly
(KLHuVhAIG, SEQ. ID NO. 13) . (ii) Ser24 to Ala24, Val48Ala49 to IleGly and Ser74 to Ala (KLHuVhAIGA, SEQ. ID NO. 14) .
(iii) Ser 24 to Ala24, VAl48Ala49 to IleGly, Ser74 to Ala and Phe79 to Tyr (KLHuVhAIGAY, SEQ. ID NO. 15) . Mutation rationale Ala24 - The loop of CDR1 is anchored by the penetration of the side chain of residue 29 into the framework. Residue 24 is one of those with which it interacts (Chothia et al . , J. Mol . Biol . 227: 799-817 (1992) ) .
Ile48Gly49 - Although both these residues are adjacent to the CDR2 hypervariable region they are far removed from the actual structural loop. Both residues are completely buried (Padlan, Mol . Immunolog. 28: 489- 498, 1991) and it was considered possible that these would effect binding via their packing interaction.
Ala74 - This residue is part of the fourth loop found at the VH antigen-binding surface and its side chain is almost completely exposed to solvent (Padlan, 1991 above) . Direct interaction of this residue with antigen could be envisaged.
Tyr79 - Like residue 74, this residue is close to the antigen-binding site and could effect binding.
The alignment of the different versions of KOL VH frameworks (SEQ. ID NOS. 7 and 12-15) with NEWM based versions (SEQ. ID NOS. 5 and 8-11) and the murine MN14 (SEQ. ID NO. 2) is shown in Figure 6. The DNA sequences and translation products of MN14HuVH and MN14HuVL are • shown in Figures 7 and 8, Orespectively (SEQ. ID NOS. 16-19, respectively) .
The humanized KLHuVH varients, such as antibody KLHuVHAIGA/HuVK, were purified and tested in a blocking assay carried out as follows. Antibodies were added at the indicated concentrations together with HRP-labeled MN-14 to a final volume of 0.1 ml. Following 30 mins. of incubation at 37CC, and washing to remove unbound antibodies, the relative affinities of the antibodies were determined from the remaining bound peroxidase activity. A shown by the assay data of Figure 9, the activity of the "reshaped" (i.e., CDR-engrafted on FR) humanized antibody was similar to that of chimeric and murine positive controls. Results obtained using supernatant fluids from cells secreting KLHuVHAIG/HuVK and KLHuVHAIGAY/HuVK antibodies suggests that these have blocking activities that are similar to that of the KLHuVHAIGA/HuVK antibody. C. REI based humanization
REI VL is described in Epp et al . , Eur. J. Biochem. , 45: 513-24 (1974). No change4s were made in REI framework to improve binding. The changes from the sequences given in Kabat et al. (1987) are: M4 to L; T39 to K: Y71 to F; L104 to V; Q105 to E: and T107 to K. These changes preexisted in the template used when grafting the MN-14 CDRs and were not made specifically to improve binding. The M4 to L change incorporates a restriction site but the remaining differences eliminate unusual residues in the REI framework. A similar framework has been referred to as a consensus of huma kappa subgroup I by Foote et al. , J. Mol . Biol . 224: 487- 9 (1992) EXAMPLE 9
Expression of MN-14 CDR-grafted Humanized Antibodies Cells that were stably transformed for expression of the MN-14 CDR-grafted human antibodies were selected in the manner described above and cloned out to establish individual producer lines. Each of the lines was assayed to determine production of the correct antibody and to assess the efficiency of production. The antibody class was determined and the anti-CEA binding affinity assessed. The best producers were further characterized for the overall amount of the antibody produced and, for the best of these, sequences were obtained from the mRNA to insure that the mutation has not occurred in the antibody genes during transfection, integration, propagation or selection.
EXAMPLE 10
Purification of MN-14 CDR-grafted Humanized Antibodies Expressed in Cell Culture
The best producer lines of the MN-14 CDR-grafted human antibody were cultured, the growth medium collected and filtered through a 0.2 micron membrane. The antibody was then purified by protein A chromatography followed by other conventional purification steps such as ion exchange and size exclusion chromotography. The cells were pelleted and from the supernatant by conventional centrifugation. The antibody was purified from the supernatant fluid as described above. EXAMPLE 11
USES OF HUMANIZED MN-14 MONOCLONAL ANTIBODY IN
DIAGNOSES A. Animal studies
The biodistribution of labeled humanized MN-14 IgG in nude mice bearing human colon cancer was determined. For radiolocalization studies, at 4-5 weeks female athymic mice (nu/nu, Harlan, Indianapolis, IN) were given s.c. 0.2 ml of a 10% suspension of LS174T human colon adenocarcinoma prepared from a xenograft serially propagated in an athymic mouse (Sharkey et al . , Cancer Res . , 50: 828-34 (1990)) . After waiting 2 weeks for tumor development, the mice were injected i.v. with 20 μCi (about 2 μg) of "'-labelled humanized MN-14 monoclonal antibodies. Groups of 4-5 mice were sacrificed at intervals thereafter, and radioactivity localized in tissues according to Sharkey et al . , above. The data of Table 2 show the % injected dose/g tissue and tumor:nontumor ratios.
The results show excellent tumor accretion of the antibody, with maximum accretion occuring within 2 days. Blood clearance of the hMN-14 antibody was more rapid than the parental mMN-14 antibody. In addition, there was higher uptake of hMN-14 by the spleen than there was of mMN-14, reflecting the fact that the former antibody is "foreign" to the mouse. Tumor:nontumor ratios were excellent. These results demonstrate that the inventive hMN-14 mAb is capable of targeting CEA-producing tumors. TABLE 2
Percent Injected Dose Per Gram Tissue (N = 4 to 5 animals)
Time Post-Injection ,31l-hMN-14 IgG
Tissue 4 hour 1 day 2 days 5 days 7 days 14 days
LS174T 1 1.8 ± 2.9 18.1 ± 14.9 32.6 ± 17.2 30.2 ± 13.4 10.6 ± 15.2 1 1.6 ± 5.6 weight 0.31 ± 0.07 0.37 ± 0.02 0.27 ± 0.08 0.31 ± 0.9 0.3 ± 0.2 0.40 ± 0.07 V".
Liver 10.8 ± 2.1 6.0 ± 3.3 2.8 ± 0.5 0.8 ± 0.4 0.5 ± 0.7 0.08 ± 0.05
Spleen 17.0 ± 4.8 10.5 ± 8.8 4.9 ± 0.4 1.3 ± 0.6 0.6 ± 0.8 0.14 ± 0.09
Kidney 7.0 ± 0.8 3.1 ± 0.7 2.3 ± 0.9 0.9 ± 0.4 0.4 ± 0.6 0.07 ± 0.04
Lungs 8.7 ± 0.4 3.7 ± 1 .4 3.7 ± 1 .4 1 .4 ± 0.6 0.6 ± 1 .0 0.1 1 ± 0.06
Blood 15.4 ± 6.6 5.5 ± 4.7 6.8 ± 4.1 2.3 ± 1.2 0.9 ± 1.8 0.14 ± 0.13
Figure imgf000038_0001
Tumor (LS1474T)/Nontumor Ratios (N = 4 to 5 animals)
Time Post-Injection ,3,l-hMN-14 IgG
Tissue 4 hour 1 day 2 days 5 days 7 days 1 days
Liver 1 .1 ± 0.3 5.0 ± 5.0 1 1 .1 ± 4.7 39.5 ± 7.5 24.6 ± 4.8 160 ± 28
Spleen 0.7 ± 0.3 4.2 ± 4.6 6.6 ± 3.4 24.7 ± 7.8 15.8 ± 4.4 91 ± 21
Kidney 1 .7 ± 0.5 5.3 ± 3.5 13.3 ± 3.6 36.1 ± 4.0 38.4 ± 12.9 173 ± 41
Lungs 1.4 ± 0.4 4.2 ± 2.2 8.2 ± 2.1 22.3 ± 2 1 25.4 ± 7.3 1 12 ± 15
Figure imgf000038_0002
Blood 0.9 ± 0.5 3.7 ± 0.9 5.4 ± 1.4 14.2 ± 2.9 26.0 ± 12.9 111 ± 35
B. Clinical Studies with 131I-labeled Humanized MN-14 IgG Patients were entered into an Institutional Review Board-approved protocol at the Center for Molecular Medicine and Immunology, Newark, NJ for a pilot investigation of the targeting and pharmacokinetic behavior of the humanized MN-14 IgG. In the case the results of which are shown in Figure 12, the male patient had colorectal cancer that had metastasized to the liver. He was injected i.v. with 131I-hMN-14 IgG (8 mCi, 0.6 mg antibody) and images were taken over a six day period. The patient was subsequently injected with an identical dose of mMN-14 IgG. The images shown in Figure 12 shaw the anterior abdominal view about 140 h after each injection. The images are adjusted to exactly the same intensity so that they are directly comparable. The results indicate that the humanized antibody is taken up by the CEA-producing tumor as well as the parental murine antibody. These experiments establish the practical utility of diagnosing human CEA-producing colon cancers with the inventive humanized MN-14 mAb.

Claims

WHAT IS CLAIMED IS:
1. A humanized monoclonal antibody, comprising the complementarity-determining regions (CDRs) of a parental murine Class III, anti-CEA monoclonal antibody engrafted to the framework regions (FRs) of a heterologous antibody, wherein said heterologous includes any species including human, wherein said humanized antibody retains the Class III, anti-CEA binding specificity of said parental murine monoclonal antibody but is less immunogenic in a human subject than is said parental murine Class III, anti-CEA monoclonal antibody.
2. A humanized monoclonal antibody of Claim 1, wherein said murine Class III, anti-CEA monoclonal antibody is the MN-14 monoclonal antibody.
3. A humanized monoclonal antibody of Claim 1, wherein said heterologous antibody is a human antibody.
4. A humanized monoclonal antibody of Claim 2, wherein:
(a) the light chain variable regions are characterized by the formula:
FRL1- CDRL1- FRL2-CDRL2-FRL3-CDRLJ-FR , wherein each FR is a different framework region of a human antibody, and each CDR is a different complementarity determining region, of the light chain of MN-14; and,
(b) the heavy chain variable regions are characterized by the formula:
FRH1-CDRHι-FRm-CDRjβ- FR_u-CDRrø-FRH4, wherein each FR is a different framework regions of a human antibody and each CDR is a different complementarity determining region of said heavy chain of MN-14.
5. A humanized monoclonal antibody according to Claim 4, wherein the amino acid sequence of : CDRLI is KASQD VGTSV A (SEQ. ID. NO. 20) ; CDRLZ is WTSTR HT (SEQ. ID. NO. 21) ; CDRU is QQYSL YRS (SEQ. ID. NO. 22) ; CDRH1 is TYWMS (SEQ. ID. NO. 23) ; CDRfβ is EIHPD SSTIN YAPSL KD (SEQ. ID NO. 24) ; and, CDRHS is LYFGF PWFAY (SEQ. ID NO. 25) .
6. A humanized monoclonal antibody according to Claim 4, wherein: FRL1 comprises a region of about 23 amino acids that occurs naturally in the FRL1 of a human antibody; FRL2 comprises a region of about 15 amino acids that occurs naturally in the FRL22 of a human antibody; FRU comprises a region of about 32 amino acids that occurs naturally in the FRL33 of a human antibody;
FRW comprises a region of about 10 amino acids that occurs naturally in the FRU of a human antibody; FRH1 comprises a region of 28-32 amino acids that occurs naturally in the FRHI of a human antibody; FRHJ comprises a region of 12-16 amino acids that occurs naturally in the FR^ of a human antibody; FRrø comprises a region of 30-34 amino acids that occurs naturally in the FRrø of a human antibody; and, FRH4 comprises a region of 9-13 amino acids that occurs naturally in the FRH4 of a human antibody.
7. A humanized monoclonal antibody according to Claim 6, wherein the amino acid sequence of:
FRL, is DIQLT QSPSS LSASV GDRVT ITC (SEQ. ID NO. 26);
FRL2 is WYQQK PGKAP KLLIY (SEQ. ID NO. 27); FRu is GVP(S or D)R FSGS(G or V) SGTDF TFTIS SLQPE DIATY
YC (SEQ. ID NO. 28) ;
FRU is FGQGT KVIEK (SEQ. ID NO. 29);
FRHι is EVQLV ESGGG WQPG RSLRL SCSSS GFDFT (SEQ. ID NO.
30) , EVQLV ESGGG WQPG RSLRL SCSAS GFDFT (SEQ. ID NO. 31) , or
QVQLQ ESGPG LVRPS QTLSL TCTSS GFDFT (SEQ. ID NO. 32) ;
FRm is WVRQA PGKGL EWVA (SEQ. ID NO. 33), WVRQA PGKGL EWIA or (SEQ. ID NO. 34) WVRQP PGRGL EWIA (SEQ. ID NO. 35);
FRH3 is RFTIS RDNSK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID
NO. 36) , RFTIS RDNAK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID NO. 37) or
RVTML RDTSK NGSFL RLSSV TAADT AVYYC AS (SEQ. ID NO. 38) ; and FRH4 is WGQGT PVTVS S (SEQ. ID NO. 39) or WGQGT TVTVS S (SEQ. ID NO. 40) ; and wherein C may be in the sulfhydryl or disulfide form.
8. A humanized monoclonal antibody according to Claim 7, wherein the amino acid sequence of: CDRL1 is KASQD VGTSV A (SEQ. ID NO. 20);
CDRU is WTSTR HT (SEQ. ID NO. 21);
CDRL3 is QQYSL YRS (SEQ. ID NO. 22);
CDRHι is TYWMS (SEQ. ID NO. 23);
CDRHZ is EIHPD SSTIN YAPSL KD (SEQ. ID NO. 24) ; and CDRrø is LYFGF PWFAY (SEQ. ID NO. 25) .
9. A humanized monoclonal antibody according to Claim 1 designated KLHuVHAIGA
10. A conjugate comprising a diagnostic or therapeutic agent bound to a murine Class III, anti-CEA, humanized monoclonal antibody, wherein the CDRs of said antibody are derived from a parental murine Class III, anti-CEA monoclonal antibody and the FRs are derived from a heterologous antibody, said heterologous referring to any species including human, wherein said humanized monoclonal antibody retains the Class III, anti-CEA binding specificity of said parental murine Class III, anti-CEA monoclonal antibody but is less immunogenic in said heterologous species than is said parental Class III, anti-CEA monoclonal antibody.
11. A conjugate according to Claim 9, wherein said parental murine Class III, anti-CEA monoclonal antibody is the MN-14 monoclonal antibody.
12. A conjugate according to Claim 10 wherein said heterologous refers to a human.
13. A conjugate according to Claim 12, wherein in said monoclonal antibody:
(a) the light chain variable regions are characterized by the formula: FRL1- CDRL1- FRLJ- CDR^- FRU- CDRU- FR , wherein each FR is a different framework region of a human antibody, and each CDR is a different complementarity determining region, of the light chain of MN-14; and,
(b) the heavy chain variable regions are characterized by the formula:
FRH1-CDRHι-FRH2-CDR-fβ-FRrø-CDRrø-FRH , wherein each FR is a different framework region of a human antibody, and each CDR is a different complementarity determining region, of the heavy chain of MN-14.
14. A conjugate according to Claim 13, wherein the amino acid sequence of : CDRL1 is KASQD VGTSV A (SEQ. ID NO. 20) ;
CDRu is WTSTR HT (SEQ. ID NO. 21) ;
CDRL3 is QQYSL YRS (SEQ. ID NO. 22) ;
CDRH1 is TYWMS (SEQ. ID NO. 23);
CDRjc is EIHPD SSTIN YAPSL KD (SEQ. ID NO. 24) ; and, CDRrø is LYFGF PWFAY (SEQ. ID NO. 25) .
15. A conjugate according to Claim 13, wherein: FR comprises a region of about 23 amino acids that occurs naturally in the FRL1 of a human antibody;
FRu comprises a region of about 15 amino acids that occurs naturally in the FRL^ of a human antibody;
FRu comprises a region of about 32 amino acids that occurs naturally in the FRL33 of a human antibody; FR comprises a region of about 10 amino acids that occurs naturally in the FR of a human antibody; FRH1 comprises a region of 28-32 amino acids that occurs naturally in the FRH, of a human antibody; FRjβ comprises a region of 12-16 amino acids that occurs naturally in the FRrø of a human antibody; FRJO comprises a region of 30-34 amino acids that occurs naturally in the FRm of a human antibody; and, FRH4 comprises a region of 9-13 amino acids that occurs naturally in the FRH4 of a human antibody.
16. A conjugate according to Claim 15, wherein the amino acid sequence of :
FR is DIQLT QSPSS LSASV GDRVT ITC (SEQ. ID NO. 26) ;
FRU is WYQQK PGKAP KLLIY (SEQ. ID NO. 27); FRU is GVP(S or D)R FSGS (G or V) SGTDF TFTIS SLQPE DIATY
YC (SEQ. ID NO. 28) ;
FR is FGQGT KVIEK (SEQ. ID NO. 29) ;
FRH1 is EVQLV ESGGG WQPG RSLRL SCSSS GFDFT (SEQ. ID NO.
30) , EVQLV ESGGG WQPG RSLRL SCSAS GFDFT (SEQ. ID NO. 31) , or
QVQLQ ESGPG LVRPS QTLSL TCTSS GFDFT (SEQ. ID NO. 32) ;
FRH-2 is WVRQA PGKGL EWVA (SEQ. ID NO. 33), WVRQA PGKGL EWIA (SEQ. ID NO. 34)or WVRQP PGRGL EWIA (SEQ. ID NO. 35);
FRHJ is RFTIS RDNSK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID
NO. 36) ,
RFTIS RDNAK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID NO.
37) or RVTML RDTSK NGSFL RLSSV TAADT AVYYC AS (SEQ. ID NO.
38) ; and
FRH4 is WGQGT PVTVS S (SEQ. ID NO. 39) or WGQGT TVTVS S
(SEQ. ID NO. 40) ; and wherein C may be in the sulfhydryl or disulfide form.
17. A conjugate according to Claim 16, wherein the amino acid sequence of:
CDRL, is KASQD VGTSV A (SEQ. ID NO. 20);
CDRU is WTSTR HT (SEQ. ID NO. 21);
CDRL-3 is QQYSL YRS (SEQ. ID NO. 22) ; CDRH1 is TYWMS (SEQ. ID NO. 23) ;
CDRm is EIHPD SSTIN YAPSL KD (SEQ. ID NO. 24); and
CDRH-J is LYFGF PWFAY (SEQ. ID NO. 25) .
18. A conjugate according to Claim 10 wherein said therapeutic agent comprises a cytotoxic agent.
19. A conjugate according to Claim 10 wherein said diagnostic reagent comprises an imaging agent.
20. A method for diagnosing or treating a patient comprising the step of administering to said patient in an effective amount for diagnosis or treatment a conjugate comprising a diagnostic or therapeutic agent as described in any one of claims 10-19, inclusive.
21. An isolated, purified chimeric DNA that encodes the CDRs of the light and heavy chains of a parental murine Class III, anti-CEA monoclonal antibody and the FRs of a heterologous antibody, whwrein said heterologous antibody can be derived from any species including human, wherein the antibody expressed by said chimeric DNA retains the Class III, anti-CEA binding specificity of said parental murine Class III, anti-CEA monoclonal antibody, but is less immunogenic in a heterologous subject than is said parental monoclonal antibody.
22. An isolated polynucleotide comprising a DNA sequence encoding the VH amino acid chain of the MN-14 monoclonal antibody as set out in Figure 1, and alleles, variants and mutations thereof.
23. A protein encoded by the DNA sequence of Claim 22.
24. An isolated polynucleotide comprising a DNA sequence encoding the VK amino acid chain of the MN-14 monoclonal antibody as set out in Figure 2, and alleles, variants and mutations thereof.
25. A protein encoded by the DNA sequence of Claim 24.
26. A vector for the expression of a humanized chimeric MN-14 heavy chain, said vector being designated pSVgptMN14MuVHHuIgGl, as described in Figure 3.
27. A vector for the expression of a humanized chimeric MN-14 kappa light chain, said vector being designated pSVhygMN14MuVKHuCK, as described in Figure 4.
28. A method for the expression of a humanized chimeric MN-14 monoclonal antibody, comprising the steps of:
(a) linearizing vectors pSVgptMN14MuVHHuIgGl and pSVhygMN14MuVKHuCK; (b) transfecting mammalian lymphoma cells with said linearized vectors;
(c) selecting said transfected cells for those expressing the gpt gene; and (d) selecting from said cells expressing said gpt gene cells secreting said humanized chimeric monoclonal antibody.
29. A vector for the expression of a reshaped humanized MN-14 variable heavy chain, designated pSVgptMN14NEWMHuIgGl, as described in Figure 3, wherein MN-14 amino acid residues at positions 24, 27, 28, 30, 71 and 94 are mutated into said variable heavy chain along with the CDRs of said MN-14 variable heavy chain.
30. A vector for the expression of a reshaped humanized MN-14 variable kappa light chain, designated pSVhgrMN14REIHuCK, as described in Figure 4, wherein MN-14 amino acid residues 24, 27, 28, 30, 71 and 94 are mutated into said variable kappa light chain along with the CDRs of said MN-14 variable kappa light chain.
31. An isolated, purified DNA according to Claim 21, wherein:
(a) said light chain is characterized by a variable region of the formula:
FRL1- CDRL1- FRu-CDRu-FRLa-CDR^-FR , wherein each FR is a different framework region of a human antibody, and each CDR is a different complementarity determining region, of the light chain of MN-14; and,
(b) the heavy chain variable regions are characterized by the formula:
FRm-CDRH1-FRjj2-CDRjβ-FRrø-CDRrø-FRμ , wherein each FR is a different framework region of a human antibody and each CDR is a different complementarity determining region of the heavy chain of MN-14.
32. An isolated, purified DNA according to Claim 31, wherein the amino acid sequence of:
CDRL1 is KASQD VGTSV A (SEQ. ID NO. 20); CDRU is WTSTR HT (SEQ. ID NO. 21) ; CDRu is QQYSL YRS (SEQ. ID NO. 22) ; CDRH1 is TYWMS (SEQ. ID NO. 23) ;
CDRm is EIHPD SSTIN YAPSL KD (SEQ. ID NO. 24) ; and, CDRH3 is LYFGF PWFAY (SEQ. ID NO. 25) .
33. An isolated, purified DNA according to Claim 31, wherein:
FRL1 comprises a region of about 23 amino acids that occurs naturally in the FRLI of a human antibody; FRu comprises a region of about 15 amino acids that occurs naturally in the FRL22 of a human antibody; FRL3 comprises a region of about 32 amino acids that occurs naturally in the FRL33 of a human antibody; FR comprises a region of about 10 amino acids that occurs naturally in the FRW of a human antibody; FRH1 comprises a region of 28-32 amino acids that occurs naturally in the FRH, of a human antibody;
FRrø comprises a region of 12-16 amino acids that occurs naturally in the FRrø of a human antibody; FRm comprises a region of 30-34 amino acids that occurs naturally in the FRH3 of a human antibody; and, FRH4 comprises a region of 9-13 amino acids that occurs naturally in the FRH4 of a human antibody.
34. An isolated, purified DNA according to Claim 33, wherein the amino acid sequence of :
FRL1 is DIQLT QSPSS LSASV GDRVT ITC (SEQ. ID NO. 26) ; FRU is WYQQK PGKAP KLLIY (SEQ. ID NO. 27) ;
FRU is GVP(S or D)R FSGS (G or V) SGTDF TFTIS SLQPE DIATY
YC6 (SEQ. ID NO. 28) ;
FR is FGQGT KVIEK (SEQ. ID NO. 29) ;
FRH1 is EVQLV ESGGG WQPG RSLRL SCSSS GFDFT (SEQ. ID NO. 30) ,
EVQLV ESGGG WQPG RSLRL SCSAS GFDFT (SEQ. ID NO. 31) , or QVQLQ ESGPG LVRPS QTLSL TCTSS GFDFT (SEQ. ID NO. 32) ; FRm is WVRQA PGKGL EWVA (SEQ. ID NO. 33),
WVRQA PGKGL EWIA (SEQ. ID NO. 34) or
WVRQP PGRGL EWIA (SEQ. ID NO. 35) ; FRH3 is RFTIS RDNSK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID NO. 36),
RFTIS RDNAK NTLFL QMDSL RPEDT GVYFC AS (SEQ. ID NO. 37) or
RVTML RDTSK NGSFL RLSSV TAADT AVYYC AS (SEQ. ID NO. 38) ; and
FRH4 is WGQGT PVTVS S (SEQ. ID NO. 39) or WGQGT TVTVS S (SEQ. ID NO. 40) ; and wherein C may be in the sulfhydryl or disulfide form.
35. An isolated, purified DNA according to Claim 34, wherein the amino acid sequence of :
CDRL1 is KASQD VGTSV A (SEQ. ID NO. 20) ; CDRLJ is WTSTR HT (SEQ. ID NO. 21) ; CDRu is QQYSL YRS (SEQ. ID NO. 22) ; CDRH1 is TYWMS (SEQ. ID NO. 23) CDRm is EIHPD SSTIN YAPSL KD (SEQ. ID NO. 24) ; and CDRHJ is LYFGF PWFAY (SEQ. ID NO. 25) .
36. Plasmid pSVgpt of Figure 3 containing a chimeric reshaped gene.
37. Plasmid PSVhyg of Figure 4 containing a chimeric gene.
38. Transformed cells expressing the chimeric gene shown in Figure 3.
39. Transformed cells expressing the chimeric gene shown in Figure 4.
PCT/US1995/011964 1994-10-05 1995-09-28 Cdr-grafted type iii anti-cea humanized mouse monoclonal antibodies WO1996011013A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA2200868A CA2200868C (en) 1994-10-05 1995-09-28 Cdr-grafted type iii anti-cea humanized mouse monoclonal antibodies
EP95935020A EP0783313B1 (en) 1994-10-05 1995-09-28 Cdr-grafted type iii anti-cea humanized mouse monoclonal antibodies
JP51258896A JP3437580B2 (en) 1994-10-05 1995-09-28 Type III anti-CEA humanized mouse monoclonal antibody grafted with CDR
DE69535755T DE69535755D1 (en) 1994-10-05 1995-09-28 CDR-EXCHANGED TYPE III ANTI-CEA HUMANIZED MONOCLONAL MOUSE ANTIBODY
AU37196/95A AU689331C (en) 1994-10-05 1995-09-28 CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/318,157 US5874540A (en) 1994-10-05 1994-10-05 CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
US08/318,157 1994-10-05

Publications (1)

Publication Number Publication Date
WO1996011013A1 true WO1996011013A1 (en) 1996-04-18

Family

ID=23236916

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/011964 WO1996011013A1 (en) 1994-10-05 1995-09-28 Cdr-grafted type iii anti-cea humanized mouse monoclonal antibodies

Country Status (8)

Country Link
US (3) US5874540A (en)
EP (3) EP0783313B1 (en)
JP (3) JP3437580B2 (en)
AT (2) ATE464906T1 (en)
CA (1) CA2200868C (en)
DE (2) DE69536072D1 (en)
ES (1) ES2307294T3 (en)
WO (1) WO1996011013A1 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997042329A1 (en) * 1996-05-04 1997-11-13 Zeneca Limited Monoclonal antibody to cea, conjugates comprising said antibody, and their therapeutic use in an adept system
WO1999043817A1 (en) * 1998-02-25 1999-09-02 The Dow Chemical Company High affinity humanized anti-cea monoclonal antibodies
EP0939653A1 (en) * 1996-03-20 1999-09-08 Immunomedics, Inc. Humanization of an anti-carcinoembryonic antigen anti-idiotype antibody and use as a tumor vaccine and for targeting applications
WO2002002783A2 (en) * 2000-07-03 2002-01-10 Gala Design, Inc. Expression vectors
WO2002002738A2 (en) * 2000-07-03 2002-01-10 Gala Design, Inc. Host cells containing multiple integrating vectors
US6417337B1 (en) * 1996-10-31 2002-07-09 The Dow Chemical Company High affinity humanized anti-CEA monoclonal antibodies
US6852510B2 (en) 2000-07-03 2005-02-08 Gala Design Inc Host cells containing multiple integrating vectors
US7803372B2 (en) 2002-10-08 2010-09-28 Immunomedics, Inc. Antibody therapy
US8222188B2 (en) 2002-03-28 2012-07-17 Catalent Pharma Solutions, Llc Antibody libraries
EP3066470A4 (en) * 2013-11-05 2017-05-31 Immunomedics, Inc. Humanized anti-ceacam5 antibody and uses thereof
US10316104B2 (en) 2011-04-29 2019-06-11 Roche Glycart Ag Immunoconjugates
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
WO2023077030A1 (en) 2021-10-29 2023-05-04 Gilead Sciences, Inc. Cd73 compounds
WO2023122615A1 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023147418A1 (en) 2022-01-28 2023-08-03 Gilead Sciences, Inc. Parp7 inhibitors
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
WO2023201268A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating tumor antigen expressing cancers
WO2023205719A1 (en) 2022-04-21 2023-10-26 Gilead Sciences, Inc. Kras g12d modulating compounds
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds

Families Citing this family (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7354587B1 (en) * 1994-07-06 2008-04-08 Immunomedics, Inc. Use of immunoconjugates to enhance the efficacy of multi-stage cascade boosting vaccines
US5798100A (en) 1994-07-06 1998-08-25 Immunomedics, Inc. Multi-stage cascade boosting vaccine
US6703488B1 (en) * 1998-01-15 2004-03-09 Center For Molecular Medicine And Immunology Antibody/receptor targeting moiety for enhanced delivery of armed ligand
US6962702B2 (en) 1998-06-22 2005-11-08 Immunomedics Inc. Production and use of novel peptide-based agents for use with bi-specific antibodies
MY133346A (en) * 1999-03-01 2007-11-30 Biogen Inc Kit for radiolabeling ligands with yttrium-90
US20020102208A1 (en) 1999-03-01 2002-08-01 Paul Chinn Radiolabeling kit and binding assay
US20030099629A1 (en) * 1999-03-11 2003-05-29 Immunomedics, Inc. Recombinant onconase and chemical conjugates and fusion proteins of recombinant onconase
US7527787B2 (en) * 2005-10-19 2009-05-05 Ibc Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US7829064B2 (en) * 1999-05-10 2010-11-09 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods
US8119101B2 (en) 1999-05-10 2012-02-21 The Ohio State University Anti-CD74 immunoconjugates and methods of use
US8383081B2 (en) * 1999-05-10 2013-02-26 Immunomedics, Inc. Anti-CD74 immunoconjugates and methods of use
US20030224415A1 (en) * 2001-06-29 2003-12-04 Gala Design, Inc. Selection free growth of host cells containing multiple integrating vectors
US20040235173A1 (en) * 2000-07-03 2004-11-25 Gala Design, Inc. Production of host cells containing multiple integrating vectors by serial transduction
CA2417185A1 (en) * 2000-07-25 2002-01-31 Shui-On Leung Multivalent target binding protein
US20020165360A1 (en) * 2000-11-30 2002-11-07 Junghans Richard P. Chimeric effector cell receptors against carcinoembryonic antigen
WO2002078638A2 (en) * 2001-03-30 2002-10-10 University Of Massachusetts Morpholino imaging and therapy
KR101027889B1 (en) * 2001-10-15 2011-04-07 아이비씨 파마슈티컬스, 인코퍼레이티드 Affinity enhancement agents
US7238785B2 (en) 2002-03-01 2007-07-03 Immunomedics, Inc. RS7 antibodies
US7591994B2 (en) 2002-12-13 2009-09-22 Immunomedics, Inc. Camptothecin-binding moiety conjugates
US9770517B2 (en) 2002-03-01 2017-09-26 Immunomedics, Inc. Anti-Trop-2 antibody-drug conjugates and uses thereof
EP1487879B1 (en) 2002-03-01 2012-12-26 Immunomedics, Inc. Bispecific antibody point mutations for enhancing rate of clearance
US20160279239A1 (en) 2011-05-02 2016-09-29 Immunomedics, Inc. Subcutaneous administration of anti-cd74 antibody for systemic lupus erythematosus and autoimmune disease
ATE477276T1 (en) 2002-03-01 2010-08-15 Immunomedics Inc INTERNALIZATION OF ANTI CD74 MONOCLONAL ANTIBODIES AND THEIR USES
US7384738B2 (en) * 2002-03-28 2008-06-10 Bremel Robert D Retrovirus-based genomic screening
AU2003241024A1 (en) * 2002-05-29 2003-12-19 Immunomedics, Inc. Methods and compositions for radioimmunotherapy of brain and cns tumors
CN100548374C (en) * 2002-10-08 2009-10-14 免疫医疗公司 Carry out therapeutic alliance with anti-CEA monoclonal antibody of III class and therapeutic agent
EP1572242B1 (en) 2002-12-13 2014-04-16 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
US8420086B2 (en) 2002-12-13 2013-04-16 Immunomedics, Inc. Camptothecin conjugates of anti-CD22 antibodies for treatment of B cell diseases
AU2004255216B2 (en) * 2003-07-01 2010-08-19 Immunomedics, Inc. Multivalent carriers of bi-specific antibodies
CA2533878A1 (en) * 2003-07-29 2005-09-22 Immunomedics, Inc. Fluorinated carbohydrate conjugates
US6878531B1 (en) * 2003-11-10 2005-04-12 Medical College Of Georgia Research Institute Method for multiple site-directed mutagenesis
US20050132544A1 (en) * 2003-12-23 2005-06-23 Jayshree Seth Split hook fastener
US20050221429A1 (en) * 2004-01-16 2005-10-06 Cardinal Health Pts, Llc Host cells containing multiple integrating vectors comprising an amplifiable marker
US8883160B2 (en) * 2004-02-13 2014-11-11 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for therapeutic and diagnostic use
US8491914B2 (en) * 2004-02-13 2013-07-23 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for delivery of interference RNA
US9550838B2 (en) 2004-02-13 2017-01-24 Ibc Pharmaceuticals, Inc. Dock-and-lock (DNL) complexes for therapeutic and diagnostic use
US20110064754A1 (en) * 2005-03-03 2011-03-17 Center For Molecular Medicine And Immunology Immunoconjugates Comprising Poxvirus-Derived Peptides and Antibodies Against Antigen-Presenting Cells for Subunit-Based Poxvirus Vaccines
US8652484B2 (en) 2004-02-13 2014-02-18 Immunomedics, Inc. Delivery system for cytotoxic drugs by bispecific antibody pretargeting
US20110020273A1 (en) * 2005-04-06 2011-01-27 Ibc Pharmaceuticals, Inc. Bispecific Immunocytokine Dock-and-Lock (DNL) Complexes and Therapeutic Use Thereof
US8435539B2 (en) * 2004-02-13 2013-05-07 Immunomedics, Inc. Delivery system for cytotoxic drugs by bispecific antibody pretargeting
US8562988B2 (en) * 2005-10-19 2013-10-22 Ibc Pharmaceuticals, Inc. Strategies for improved cancer vaccines
US9481878B2 (en) 2004-02-13 2016-11-01 Immunomedics, Inc. Compositions and methods of use of immunotoxins comprising ranpirnase (Rap) show potent cytotoxic activity
US8551480B2 (en) 2004-02-13 2013-10-08 Immunomedics, Inc. Compositions and methods of use of immunotoxins comprising ranpirnase (Rap) show potent cytotoxic activity
US20060127311A1 (en) * 2004-11-17 2006-06-15 Duke University Radiation dosimetry and blocking antibodies and methods and uses therefor in the treatment of cancer
US10058621B2 (en) 2015-06-25 2018-08-28 Immunomedics, Inc. Combination therapy with anti-HLA-DR antibodies and kinase inhibitors in hematopoietic cancers
US20160355591A1 (en) 2011-05-02 2016-12-08 Immunomedics, Inc. Subcutaneous anti-hla-dr monoclonal antibody for treatment of hematologic malignancies
US9707302B2 (en) 2013-07-23 2017-07-18 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US8349332B2 (en) 2005-04-06 2013-01-08 Ibc Pharmaceuticals, Inc. Multiple signaling pathways induced by hexavalent, monospecific and bispecific antibodies for enhanced toxicity to B-cell lymphomas and other diseases
US8475794B2 (en) 2005-04-06 2013-07-02 Ibc Pharmaceuticals, Inc. Combination therapy with anti-CD74 antibodies provides enhanced toxicity to malignancies, Autoimmune disease and other diseases
US20060257317A1 (en) * 2005-05-04 2006-11-16 Duke University Combination therapy in the treatment of cancer
CN101534865A (en) * 2005-10-19 2009-09-16 Ibc药品公司 Methods and compositions for generating bioactive assemblies of increased complexity and uses
US8377440B2 (en) 2006-12-01 2013-02-19 Selexys Pharmaceuticals Corporation Anti-P-selectin antibodies and methods of using the same to treat inflammatory diseases
BRPI0911652A2 (en) 2008-04-11 2015-08-04 Merrimack Pharmaceuticals Inc Human serum albumin binders and their conjugates
EP2326346A4 (en) * 2008-08-20 2012-06-13 Ibc Pharmaceuticals Inc Dock-and-lock (dnl) vaccines for cancer therapy
EP4083072A1 (en) 2008-10-06 2022-11-02 Minerva Biotechnologies Corporation Muc1* antibodies
US8927694B2 (en) * 2008-11-18 2015-01-06 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
EP3939617A1 (en) 2009-02-13 2022-01-19 Immunomedics, Inc. Conjugates with an intracellularly-cleavable linkage
CA2774260C (en) 2009-09-16 2018-10-09 Immunomedics, Inc. Class i anti-cea antibodies and uses thereof
IN2012DN03354A (en) 2009-12-02 2015-10-23 Immunomedics Inc
EP2523680A4 (en) * 2010-01-11 2013-06-19 Ct Molecular Med & Immunology Enhanced cytotoxicity of anti-cd74 and anti-hla-dr antibodies with interferon-gamma
CN101928347B (en) * 2010-05-05 2013-03-27 上海海抗中医药科技发展有限公司 Anti-carcinoembryonic-antigen (CEA) antibody and application thereof
UA118646C2 (en) 2010-10-13 2019-02-25 Янссен Байотек, Інк. Human oncostatin m antibodies and methods of use
JP2012171088A (en) * 2011-02-24 2012-09-10 Olympus Corp Master operation input device, and master-slave manipulator
EP2681244B1 (en) * 2011-03-02 2017-11-29 Roche Glycart AG Cea antibodies
CN107115526A (en) 2011-05-02 2017-09-01 免疫医疗公司 The ultrafiltration concentration of the antibody for the allograft selection applied for small size
WO2013085893A1 (en) 2011-12-05 2013-06-13 Immunomedics, Inc. Therapeutic use of anti-cd22 antibodies for inducing trogocytosis
US9757458B2 (en) 2011-12-05 2017-09-12 Immunomedics, Inc. Crosslinking of CD22 by epratuzumab triggers BCR signaling and caspase-dependent apoptosis in hematopoietic cancer cells
CA2874864C (en) 2012-08-14 2023-02-21 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
US20150202287A1 (en) 2012-08-30 2015-07-23 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-erbb3 agents
US10137196B2 (en) 2012-12-13 2018-11-27 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity
US10744129B2 (en) 2012-12-13 2020-08-18 Immunomedics, Inc. Therapy of small-cell lung cancer (SCLC) with a topoisomerase-I inhibiting antibody-drug conjugate (ADC) targeting Trop-2
WO2015012904A2 (en) 2012-12-13 2015-01-29 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
US10206918B2 (en) 2012-12-13 2019-02-19 Immunomedics, Inc. Efficacy of anti-HLA-DR antiboddy drug conjugate IMMU-140 (hL243-CL2A-SN-38) in HLA-DR positive cancers
US9492566B2 (en) 2012-12-13 2016-11-15 Immunomedics, Inc. Antibody-drug conjugates and uses thereof
CA2884313C (en) 2012-12-13 2023-01-03 Immunomedics, Inc. Dosages of immunoconjugates of antibodies and sn-38 for improved efficacy and decreased toxicity
US9931417B2 (en) 2012-12-13 2018-04-03 Immunomedics, Inc. Antibody-SN-38 immunoconjugates with a CL2A linker
US10413539B2 (en) 2012-12-13 2019-09-17 Immunomedics, Inc. Therapy for metastatic urothelial cancer with the antibody-drug conjugate, sacituzumab govitecan (IMMU-132)
EP2774930A1 (en) 2013-03-07 2014-09-10 Aptenia S.R.L. Metallocene compounds and labeled molecules comprising the same for in vivo imaging.
US11253606B2 (en) 2013-07-23 2022-02-22 Immunomedics, Inc. Combining anti-HLA-DR or anti-Trop-2 antibodies with microtubule inhibitors, PARP inhibitors, Bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
MX2016010683A (en) 2014-02-21 2017-05-11 Ibc Pharmaceuticals Inc Disease therapy by inducing immune response to trop-2 expressing cells.
CA2935748A1 (en) 2014-02-25 2015-09-03 Immunomedics, Inc. Humanized rfb4 anti-cd22 antibody
KR20220044377A (en) 2014-04-03 2022-04-07 아이쥐엠 바이오사이언스 인코포레이티드 Modified j-chain
CA2953567C (en) 2014-06-24 2023-09-05 Immunomedics, Inc. Anti-histone therapy for vascular necrosis in severe glomerulonephritis
CN107073135B (en) 2014-09-08 2023-06-06 鲁塔纳有限公司 Constructs for delivery of molecules into the cytoplasm of cells
CN106999517A (en) 2014-10-07 2017-08-01 免疫医疗公司 The new adjuvant purposes of antibody drug conjugate
WO2016130726A1 (en) 2015-02-10 2016-08-18 Minerva Biotechnologies Corporation Humanized anti-muc1* antibodies
CN107428837A (en) 2015-04-22 2017-12-01 免疫医疗公司 Circulate separation, detection, diagnosis and/or the identification of the positive cancer cells of TROP 2
PL3313443T3 (en) 2015-06-25 2023-11-06 Immunomedics, Inc. Combining anti-hla-dr or anti-trop-2 antibodies with microtubule inhibitors, parp inhibitors, bruton kinase inhibitors or phosphoinositide 3-kinase inhibitors significantly improves therapeutic outcome in cancer
US10195175B2 (en) 2015-06-25 2019-02-05 Immunomedics, Inc. Synergistic effect of anti-Trop-2 antibody-drug conjugate in combination therapy for triple-negative breast cancer when used with microtubule inhibitors or PARP inhibitors
EP3316885B1 (en) 2015-07-01 2021-06-23 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
ES2819870T3 (en) 2015-09-30 2021-04-19 Igm Biosciences Inc Modified J-chain binding molecules
CN108463245A (en) 2015-09-30 2018-08-28 Igm生物科学有限公司 The binding molecule of J chains with modification
US20170224837A1 (en) 2016-02-10 2017-08-10 Immunomedics, Inc. Combination of abcg2 inhibitors with sacituzumab govitecan (immu-132) overcomes resistance to sn-38 in trop-2 expressing cancers
EP3448260A4 (en) 2016-04-27 2019-10-09 Immunomedics, Inc. Efficacy of anti-trop-2-sn-38 antibody drug conjugates for therapy of tumors relapsed/refractory to checkpoint inhibitors
US10918734B2 (en) 2017-03-27 2021-02-16 Immunomedics, Inc. Treatment of high Trop-2 expressing triple negative breast cancer (TNBC) with sacituzumab govitecan (IMMU-132) overcomes homologous recombination repair (HRR) rescue mediated by Rad51
US10799597B2 (en) 2017-04-03 2020-10-13 Immunomedics, Inc. Subcutaneous administration of antibody-drug conjugates for cancer therapy
WO2020064847A1 (en) 2018-09-26 2020-04-02 Ascendis Pharma A/S Degradable hyaluronic acid hydrogels
EP3983082A1 (en) 2019-06-13 2022-04-20 Bolt Biotherapeutics, Inc. Aminobenzazepine compounds, immunoconjugates, and uses thereof
AR119382A1 (en) 2019-07-12 2021-12-15 Hoffmann La Roche PRE-TARGETING ANTIBODIES AND METHODS OF USE
WO2021046112A1 (en) 2019-09-03 2021-03-11 Bolt Biotherapeutics, Inc. Aminoquinoline compounds, immunoconjugates, and uses thereof
CA3152601A1 (en) 2019-09-30 2021-04-08 Bolt Biotherapeutics, Inc. Amide-linked, aminobenzazepine immunoconjugates, and uses thereof
BR112022007719A2 (en) 2019-10-25 2022-07-12 Bolt Biotherapeutics Inc IMMUNOCONJUGATES, BINDING COMPOUND, PHARMACEUTICAL COMPOSITION, METHOD TO TREAT CANCER, USE OF AN IMMUNOCONJUGATE AND METHOD OF PREPARING AN IMMUNOCONJUGATE
CN115996756A (en) 2020-05-08 2023-04-21 博尔特生物治疗药物有限公司 Elastase substrate peptide linker immunoconjugates and uses thereof
WO2022010797A2 (en) 2020-07-07 2022-01-13 Bionecure Therapeutics, Inc. Novel maytansinoids as adc payloads and their use for the treatment of cancer
CN115803010A (en) 2020-07-09 2023-03-14 豪夫迈·罗氏有限公司 Concentrated protein compositions, their preparation and use
CR20230076A (en) 2020-07-10 2023-03-13 Hoffmann La Roche Antibodies which bind to cancer cells and target radionuclides to said cells
IL300316A (en) 2020-08-13 2023-04-01 Bolt Biotherapeutics Inc Pyrazoloazepine immunoconjugates, and uses thereof
CA3188862A1 (en) 2020-08-20 2022-02-24 Carl Alexander Kamb Compositions and methods for treating mesothelin positive cancers
AU2021329375A1 (en) 2020-08-20 2023-04-20 A2 Biotherapeutics, Inc. Compositions and methods for treating ceacam positive cancers
US20220195066A1 (en) 2020-12-11 2022-06-23 Bolt Biotherapeutics, Inc. Anti-cea immunoconjugates, and uses thereof
JP2024501453A (en) 2020-12-11 2024-01-12 ボルト バイオセラピューティクス、インコーポレーテッド Anti-CEA immunoconjugates and their uses
WO2022152656A1 (en) 2021-01-12 2022-07-21 F. Hoffmann-La Roche Ag Split antibodies which bind to cancer cells and target radionuclides to said cells
KR20230131205A (en) 2021-01-13 2023-09-12 에프. 호프만-라 로슈 아게 combination therapy
EP4313161A1 (en) 2021-03-26 2024-02-07 Bolt Biotherapeutics, Inc. 2-amino-4-carboxamide-benzazepine immunoconjugates, and uses thereof
EP4313162A1 (en) 2021-03-26 2024-02-07 Bolt Biotherapeutics, Inc. 2-amino-4-carboxamide-benzazepine immunoconjugates, and uses thereof
EP4342497A1 (en) 2021-05-10 2024-03-27 Kawasaki Institute of Industrial Promotion Antibody having reduced binding affinity for antigen
WO2023016488A1 (en) 2021-08-13 2023-02-16 昆山新蕴达生物科技有限公司 Microtubule inhibitor-based antibody-drug conjugate
CN117881431A (en) 2021-08-24 2024-04-12 昆山新蕴达生物科技有限公司 Antibody-conjugated drug conjugated by cleavable linker
WO2023076599A1 (en) 2021-10-29 2023-05-04 Bolt Biotherapeutics, Inc. Tlr agonist immunoconjugates with cysteine-mutant antibodies, and uses thereof
WO2023180489A1 (en) 2022-03-23 2023-09-28 Synaffix B.V. Antibody-conjugates for targeting of tumours expressing carcinoembyronic antigen

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0323805A2 (en) * 1988-01-05 1989-07-12 Buchegger, Franz Novel antibodies
WO1992001059A1 (en) * 1990-07-05 1992-01-23 Celltech Limited Cdr grafted anti-cea antibodies and their production

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5156840A (en) 1982-03-09 1992-10-20 Cytogen Corporation Amine-containing porphyrin derivatives
US4818709A (en) * 1983-01-21 1989-04-04 Primus Frederick J CEA-family antigens, Anti-CEA antibodies and CEA immunoassay
US5618920A (en) * 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
US5057313A (en) 1986-02-25 1991-10-15 The Center For Molecular Medicine And Immunology Diagnostic and therapeutic antibody conjugates
GB8607679D0 (en) * 1986-03-27 1986-04-30 Winter G P Recombinant dna product
AU625613B2 (en) * 1988-01-05 1992-07-16 Novartis Ag Novel chimeric antibodies
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5081235A (en) * 1989-07-26 1992-01-14 City Of Hope Chimeric anti-cea antibody
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
DE69233254T2 (en) * 1991-06-14 2004-09-16 Genentech, Inc., South San Francisco Humanized Heregulin antibody
US5443953A (en) 1993-12-08 1995-08-22 Immunomedics, Inc. Preparation and use of immunoconjugates
US5798100A (en) * 1994-07-06 1998-08-25 Immunomedics, Inc. Multi-stage cascade boosting vaccine
WO1996004925A1 (en) * 1994-08-12 1996-02-22 Immunomedics, Inc. Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells
IL115177A0 (en) * 1994-09-16 1995-12-31 Immunomedics Inc Phosphorus-32 labeling of antibodies for cancer therapy
US7321026B2 (en) * 2001-06-27 2008-01-22 Skytech Technology Limited Framework-patched immunoglobulins

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0323805A2 (en) * 1988-01-05 1989-07-12 Buchegger, Franz Novel antibodies
WO1992001059A1 (en) * 1990-07-05 1992-01-23 Celltech Limited Cdr grafted anti-cea antibodies and their production

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6730300B2 (en) 1996-03-20 2004-05-04 Immunomedics, Inc. Humanization of an anti-carcinoembryonic antigen anti-idiotype antibody and use as a tumor vaccine and for targeting applications
US7348419B2 (en) 1996-03-20 2008-03-25 Immunomedics, Inc. Humanization of an anti-carcinoembryonic antigen anti-idiotype antibody as a tumor vaccine and for targeting applications
EP0939653A1 (en) * 1996-03-20 1999-09-08 Immunomedics, Inc. Humanization of an anti-carcinoembryonic antigen anti-idiotype antibody and use as a tumor vaccine and for targeting applications
EP0939653A4 (en) * 1996-03-20 2001-02-14 Immunomedics Inc Humanization of an anti-carcinoembryonic antigen anti-idiotype antibody and use as a tumor vaccine and for targeting applications
US6903203B2 (en) 1996-05-04 2005-06-07 Astrazeneca Uk Ltd. Monoclonal antibody to CEA, conjugates comprising said antibody and their therapeutic use
US6277599B1 (en) 1996-05-04 2001-08-21 Zeneca Limited Anti-CEA antibody designated 806.077, hybridoma and method of manufacture
WO1997042329A1 (en) * 1996-05-04 1997-11-13 Zeneca Limited Monoclonal antibody to cea, conjugates comprising said antibody, and their therapeutic use in an adept system
US6417337B1 (en) * 1996-10-31 2002-07-09 The Dow Chemical Company High affinity humanized anti-CEA monoclonal antibodies
WO1999043817A1 (en) * 1998-02-25 1999-09-02 The Dow Chemical Company High affinity humanized anti-cea monoclonal antibodies
WO2002002783A2 (en) * 2000-07-03 2002-01-10 Gala Design, Inc. Expression vectors
WO2002002738A2 (en) * 2000-07-03 2002-01-10 Gala Design, Inc. Host cells containing multiple integrating vectors
WO2002002738A3 (en) * 2000-07-03 2002-05-30 Gala Design Inc Host cells containing multiple integrating vectors
WO2002002783A3 (en) * 2000-07-03 2003-01-16 Gala Design Inc Expression vectors
US6852510B2 (en) 2000-07-03 2005-02-08 Gala Design Inc Host cells containing multiple integrating vectors
US7378273B2 (en) 2000-07-03 2008-05-27 Gala Design, Inc. Expression vectors
US8222188B2 (en) 2002-03-28 2012-07-17 Catalent Pharma Solutions, Llc Antibody libraries
US8216572B2 (en) 2002-10-08 2012-07-10 Immunomedics, Inc. Antibody therapy
US8062636B2 (en) 2002-10-08 2011-11-22 Immunomedics, Inc. Antibody therapy
US7919090B2 (en) 2002-10-08 2011-04-05 Immunomedics, Inc. Antibody therapy
US7803372B2 (en) 2002-10-08 2010-09-28 Immunomedics, Inc. Antibody therapy
US8535676B2 (en) 2002-10-08 2013-09-17 Immunomedics, Inc. Antibody therapy
US8778342B2 (en) 2002-10-08 2014-07-15 Immunomedics, Inc. Antibody therapy
US10316104B2 (en) 2011-04-29 2019-06-11 Roche Glycart Ag Immunoconjugates
US11130822B2 (en) 2011-04-29 2021-09-28 Roche Glycart Ag Immunoconjugates
EP3066470A4 (en) * 2013-11-05 2017-05-31 Immunomedics, Inc. Humanized anti-ceacam5 antibody and uses thereof
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
WO2023077030A1 (en) 2021-10-29 2023-05-04 Gilead Sciences, Inc. Cd73 compounds
WO2023122615A1 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023147418A1 (en) 2022-01-28 2023-08-03 Gilead Sciences, Inc. Parp7 inhibitors
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023178181A1 (en) 2022-03-17 2023-09-21 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
WO2023201268A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating tumor antigen expressing cancers
WO2023205719A1 (en) 2022-04-21 2023-10-26 Gilead Sciences, Inc. Kras g12d modulating compounds
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds

Also Published As

Publication number Publication date
US5874540A (en) 1999-02-23
AU3719695A (en) 1996-05-02
EP1736163B1 (en) 2010-04-21
EP0783313A4 (en) 2005-01-12
EP1736163A1 (en) 2006-12-27
ES2307294T3 (en) 2008-11-16
ATE464906T1 (en) 2010-05-15
JP3437580B2 (en) 2003-08-18
DE69536072D1 (en) 2010-06-02
EP0783313B1 (en) 2008-05-14
JPH10508466A (en) 1998-08-25
US6676924B2 (en) 2004-01-13
EP1967528A3 (en) 2008-12-17
ATE395068T1 (en) 2008-05-15
EP0783313A1 (en) 1997-07-16
CA2200868C (en) 2010-04-13
US20020018750A1 (en) 2002-02-14
DE69535755D1 (en) 2008-06-26
JP2002218990A (en) 2002-08-06
JP2010075190A (en) 2010-04-08
EP1967528A2 (en) 2008-09-10
CA2200868A1 (en) 1996-04-18
US20050089538A1 (en) 2005-04-28
JP5183611B2 (en) 2013-04-17
AU689331B2 (en) 1998-03-26
EP1967528B1 (en) 2015-03-04

Similar Documents

Publication Publication Date Title
US5874540A (en) CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
US6417337B1 (en) High affinity humanized anti-CEA monoclonal antibodies
EP0771208B1 (en) Immunoconjugates and humanized antibodies specific for b-cell lymphoma and leukemia cells
JP3904238B2 (en) Glycosylated humanized B cell specific antibody
US6307026B1 (en) Humanized antibodies directed against A33 antigen
AU675268B2 (en) Humanised antibodies directed against A33 antigen
US5993813A (en) Family of high affinity, modified antibodies for cancer treatment
US6051225A (en) Family of high affinity, modified antibodies for cancer treatment
US5645817A (en) Granulocyte-binding antibody constructs, their preparation and use
AU6338898A (en) High affinity humanized anti-cea monoclonal antibodies
AU689331C (en) CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
AU723073B2 (en) CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LT LU LV MD MG MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TT UA UG UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2200868

Country of ref document: CA

Ref country code: CA

Ref document number: 2200868

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1995935020

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995935020

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642